AU2004263823B2 - Prostate stem cell antigen (PSCA) variants and subsequences thereof - Google Patents

Prostate stem cell antigen (PSCA) variants and subsequences thereof Download PDF

Info

Publication number
AU2004263823B2
AU2004263823B2 AU2004263823A AU2004263823A AU2004263823B2 AU 2004263823 B2 AU2004263823 B2 AU 2004263823B2 AU 2004263823 A AU2004263823 A AU 2004263823A AU 2004263823 A AU2004263823 A AU 2004263823A AU 2004263823 B2 AU2004263823 B2 AU 2004263823B2
Authority
AU
Australia
Prior art keywords
psca
protein
cell
polynucleotide
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
AU2004263823A
Other versions
AU2004263823A1 (en
Inventor
Pia M. Challita-Eid
Wangmao Ge
Aya Jakobovits
Arthur B. Raitano
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agensys Inc
Original Assignee
Agensys Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agensys Inc filed Critical Agensys Inc
Publication of AU2004263823A1 publication Critical patent/AU2004263823A1/en
Application granted granted Critical
Publication of AU2004263823B2 publication Critical patent/AU2004263823B2/en
Priority to AU2008258185A priority Critical patent/AU2008258185B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/58Prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464493Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; Prostatic acid phosphatase [PAP]; Prostate-specific G-protein-coupled receptor [PSGR]
    • A61K39/464494Prostate specific antigen [PSA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1051Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from breast, e.g. the antibody being herceptin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1054Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from lung
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1057Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from liver or pancreas
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/106Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from kidney or bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1072Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from the reproductive system, e.g. ovaria, uterus, testes or prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Urology & Nephrology (AREA)
  • Optics & Photonics (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Reproductive Health (AREA)
  • Toxicology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Zoology (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)

Abstract

PSCA and its encoded protein, and variants thereof, are described wherein PSCA exhibits tissue specific expression in normal adult tissue, and is aberrantly expressed in the cancers listed in Table I. Consequently, PSCA provides a diagnostic, prognostic, propylactic and/or therapeutic target for cancer. The PSCA gene or fragment thereof, or its encoded protein, or variants thereof, or a fragment thereof, can be used to elicit a humoral or cellular immune response; antibodies or T cells reactive with PSCA can be used in active or passive immunization.

Description

WO 2005/014780 PCT/US2004/017231 PROSTATE STEM CELL ANTIGEN (PSCA) VARIANTS AND SUBSEQUENCES THEREOF Technical Field [0001] The invention described herein relates to genes and their encoded proteins, termed PSCA, expressed in certain cancers, and to diagnostic and therapeutic methods and compositions useful in the management of cancers that express PSCA.
Backcground Art [0002] Cancer is the second leading cause of human death next to coronary disease. Worldwide, millions of people die from cancer every year. In the United States alone, as reported by the American Cancer Society, cancer causes the death of well over a half-million people annually, with over 1.2 million new cases diagnosed per year. While deaths from heart disease have been declining significantly, those resulting from cancer generally are on the rise. In the early part of the next century, cancer is predicted to become the leading cause of death.
[0003] Worldwide, several cancers stand out as the leading killers. In particular, carcinomas of the lung, prostate, breast, colon, pancreas, and ovary represent the primary causes of cancer death. These and virtually all other carcinomas share a common lethal feature. With very few exceptions, metastatic disease from a carcinoma is fatal. Moreover, even for those cancer patients who initially survive their primary cancers, common experience has shown that their lives are dramatically altered. Many cancer patients experience strong anxieties driven by the awareness of the potential for recurrence or treatment failure. Many cancer patients experience physical debilitations following treatment. Furthermore, many cancer patients experience a recurrence.
[0004] Worldwide, prostate cancer is the fourth most prevalent cancer in men. In North America and Northern Europe, it is by far the most common cancer in males and is the second leading cause of cancer death in men. In the United States alone, well over 30,000 men die annually of this disease second only to lung cancer. Despite the magnitude of these figures, there is still no effective treatment for metastatic prostate cancer. Surgical prostatectomy, radiation therapy, hormone ablation therapy, surgical castration and chemotherapy continue to be the main treatment modalities. Unfortunately, these treatments are ineffective for many and are often associated with undesirable consequences.
[0005] On the diagnostic front, the lack of a prostate tumor marker that can accurately detect early-stage, localized tumors remains a significant limitation in the diagnosis and management of this disease. Although the serum prostate specific antigen (PSA) assay has been a very useful tool, however its specificity and general utility is widely regarded as lacking in several important respects.
[0006] Progress in identifying additional specific markers for prostate cancer has been improved by the generation of prostate cancer xenografts that can recapitulate different stages of the disease in mice. The LAPC (Los Angeles Prostate Cancer) xenografts are prostate cancer xenografts that have survived passage in severe combined immune deficient (SCID) mice and have exhibited the capacity to mimic the transition from androgen dependence to androgen independence (Klein etaL, 1997, Nat. Med. 3:402). More recently identified prostate cancer markers include PCTA-1 (Su et al., 1996, Proc. Natl. Acad. Sci. USA 93: 7252), prostate-specific membrane (PSM) antigen (Pinto et aL, Clin Cancer Res 1996 Sep 2 1445-51), STEAP (Hubert, etaL, Proc Natl Acad Sci U S A. 1999 Dec 7; 96(25): 14523-8) and prostate stem cell antigen (PSCA) (Reiter et 1998, Proc,. Natl. Acad. Sci. USA 95: 1735).
WO 2005/014780 PCT/LS2004/017231 [0007] While previously identified markers such as PSA, PSM, PCTA and PSCA have facilitated efforts to diagnose and treat prostate cancer, there is need for the identification of additional markers and therapeutic targets for prostate and related cancers in order to further improve diagnosis and therapy.
[0008] Renal cell carcinoma (RCG) accounts for approximately 3 percent of adult malignancies. Once adenomas reach a diameter of 2 to 3 cm, malignant potential exists. In the adult, the two principal malignant renal tumors are renal cell adenocarcinoma and transitional cell carcinoma of the renal pelvis or ureter. The incidence of renal cell adenocarcinoma is estimated at more than 29,000 cases in the United States, and more than 11,600 patients died of this disease in 1998. Transitional cell carcinoma is less frequent, with an incidence of approximately 500 cases per year in the United States.
[0009] Surgery has been the primary therapy for renal cell adenocarcinoma for many decades. Until recently, metastatic disease has been refractory to any systemic therapy. With recent developments in systemic therapies, particularly immunotherapies, metastatic renal cell carcinoma may be approached aggressively in appropriate patients with a possibility of durable responses. Nevertheless, there is a remaining need for effective therapies for these patients.
[0010] Of all new cases of cancer in the United States, bladder cancer represents approximately 5 percent in men (fifth most common neoplasm) and 3 percent in women (eighth most common neoplasm). The incidence is increasing slowly, concurrent with an increasing older population. In 1998, there was an estimated 54,500 cases, including 39,500 in men and 15,000 in women. The age-adjusted incidence in the United States is 32 per 100,000 for men and eight per 100,000 in women. The historic male/female ratio of 3:1 may be decreasing related to smoking patterns in women. There were an estimated 11,000 deaths from bladder cancer in 1998 (7,800 in men and 3,900 in women). Bladder cancer incidence and mortality strongly increase with age and will be an increasing problem as the population becomes more elderly.
[0011 IVost bladder cancers recur in the bladder. Bladder cancer is managed with a combination of transurethral resection of the bladder (TUR) and intravesical chemotherapy or immunotherapy. The multifocal and recurrent nature of bladder cancer points out the limitations of TUR. Most muscle-invasive cancers are not cured by TUR alone. Radical cystectomy and urinary diversion is the most effective means to eliminate the cancer but carry an undeniable impact on urinary and sexual function. There continues to be a significant need for treatment modalities that are beneficial for bladder cancer patients.
[0012] An estimated 130,200 cases of colorectal cancer occurred in 2000 in the United States, including 93,800 cases of colon cancer and 36,400 of rectal cancer. Colorectal cancers are the third most common cancers in men and women. Incidence rates declined significantly during 1992-1996 per year). Research suggests that these declines have been due to increased screening and polyp removal, preventing progression of polyps to invasive cancers. There were an estimated 56,300 deaths (47,700 from colon cancer, 8,600 from rectal cancer) in 2000, accounting for about 11% of all U.S. cancer deaths.
[0013] At present, surgery is the most common form of therapy for colorectal cancer, and for cancers that have not spread, it is frequently curative. Chemotherapy, or chemotherapy plus radiation, is given before or after surgery to most patients whose cancer has deeply perforated the bowel wall or has spread to the lymph nodes. A permanent colostomy (creation of an abdominal opening for elimination of body wastes) is occasionally needed for colon cancer and is infrequently required for rectal cancer. There continues to be a need for effective diagnostic and treatment modalities for colorectal cancer.
WO 2005/014780 PCT/LS2004/017231 [0014] There were an estimated 164,100 new cases of lung and bronchial cancer in 2000, accounting for 14% of all U.S. cancer diagnoses. The incidence rate of lung and bronchial cancer is declining significantly in men, from a high of 86.5 per 100,000 in 1984 to 70.0 in 1996. In the 1990s, the rate of increase among women began to slow. In 1996, the incidence rate in women was 42.3 per 100,000.
[0015] Lung and bronchial cancer caused an estimated 156,900 deaths in 2000, accounting for 28% of all cancer deaths. During 1992-1996, mortality from lung cancer declined significantly among men per year) while rates for women were still significantly increasing per year). Since 1987, more women have died each year of lung cancer than breast cancer, which, for over 40 years, was the major cause of cancer death in women. Decreasing lung cancer incidence and mortality rates most likely resulted from decreased smoking rates over the previous 30 years; however, decreasing smoking patterns among women lag behind those of men. Of concern, although the declines in adult tobacco use have slowed, tobacco use in youth is increasing again.
[0016] Treatment options for lung and bronchial cancer are determined by the type and stage of the cancer and include surgery, radiation therapy, and chemotherapy. For many localized cancers, surgery is usually the treatment of choice. Because the disease has usually spread by the time it is discovered, radiation therapy and chemotherapy are often needed in combination with surgery. Chemotherapy alone or combined with radiation is the treatment of choice for small cell lung cancer; on this regimen, a large percentage of patients experience remission, which in some cases is long lasting.
There is however, an ongoing need for effective treatment and diagnostic approaches for lung and bronchial cancers.
[0017] An estimated 182,800 new invasive cases of breast cancer were expected to occur among women in the United States during 2000. Additionally, about 1,400 new cases of breast cancer were expected to be diagnosed in men in 2000. After increasing about 4% per year in the 1980s, breast cancer incidence rates in women have leveled off in the 1990s to about 110.6 cases per 100,000.
[0018] In the U.S. alone, there were an estimated 41,200 deaths (40,800 women, 400 men) in 2000 due to breast cancer. Breast cancer ranks second among cancer deaths in women. According to the most recent data, mortality rates declined significantly during 1992-1996 with the largest decreases in younger women, both white and black. These decreases were probably the result of earlier detection and improved treatment.
[0019] Taking into account the medical circumstances and the patient's preferences, treatment of breast cancer may involve lumpectomy (local removal of the tumor) and removal of the lymph nodes under the arm; mastectomy (surgical removal of the breast) and removal of the lymph nodes under the arm; radiation therapy; chemotherapy; or hormone therapy. Often, two or more methods are used in combination. Numerous studies have shown that, for early stage disease, long-term survival rates after lumpectomy plus radiotherapy are similar to survival rates after modified radical mastectomy. Significant advances in reconstruction techniques provide several options for breast reconstruction after mastectomy. Recently, such reconstruction has been done at the same time as the mastectomy.
[0020] Local excision of ductal carcinoma in situ (DCIS) with adequate amounts of surrounding normal breast tissue may prevent the local recurrence of the DCIS. Radiation to the breast and/or tamoxifen may reduce the chance of DCIS occurring in the remaining breast tissue. This is important because DCIS, if left untreated, may develop into invasive breast cancer. Nevertheless, there are serious side effects or sequelae to these treatments. There is, therefore, a need for efficacious breast cancer treatments.
[0021] There were an estimated 23,100 new cases of ovarian cancer in the United States in 2000. It accounts for 4% of all cancers among women and ranks second among gynecologic cancers. During 1992-1996, ovarian cancer 00 incidence rates were significantly declining. Consequent to ovarian cancer, there were ;an estimated 14,000 deaths in 2000. Ovarian cancer causes more deaths than any other cancer of the female reproductive system.
N, [0022] Surgery, radiation therapy, and chemotherapy are treatment options for ovarian cancer. Surgery usually includes the removal of one or both ovaries, the Cc fallopian tubes (salpingo-oophorectomy), and the uterus (hysterectomy). In some very 00 early tumors, only the involved ovary will be removed, especially in young women M who wish to have children. In advanced disease, an attempt is made to remove all intraabdominal disease to enhance the effect of chemotherapy. There continues to be an O 10 important need for effective treatment options for ovarian cancer.
NI [0023] There were an estimated 28,300 new cases of pancreatic cancer in the United States in 2000. Over the past 20 years, rates of pancreatic cancer have declined in men. Rates among women have remained approximately constant but may be beginning to decline. Pancreatic cancer caused an estimated 28,200 deaths in 2000 in the United States. Over the past 20 years, there has been a slight but significant decrease in mortality rates among men (about-0.9% per year) while rates have increased slightly among women.
10024] Surgery, radiation therapy, and chemotherapy are treatment options for pancreatic cancer. These treatment options can extend survival and/or relieve symptoms in many patients but are not likely to produce a cure for most. There is a significant need for additional therapeutic and diagnostic options for pancreatic cancer.
Disclosure of the Invention [00251 The present invention relates to a gene, designated PSCA, that has now been found to be over-expressed in the cancer(s) listed in Table I. Northern blot expression analysis of PSCA gene expression in normal tissues shows a restricted expression pattern in adult tissues. The nucleotide (Figure 2) and amino acid (Figure 2, and Figure 3) sequences of PSCA are provided. The tissue-related profile of PSCA in normal adult tissues, combined with the over-expression observed in the tissues listed in Table I, shows that PSCA is aberrantly over-expressed in at least some cancers, and thus serves as a useful diagnostic, prophylactic, prognostic, and/or therapeutic target for cancers of the tissue(s) such as those listed in Table I.
[0025A] The present invention further provides an isolated polynucleotide that encodes a PSCA protein, wherein the polynucleotide is selected from the group consisting of: 00
O
a polynucleotide comprising the sequence of SEQ ID NO:6537, from Snucleotide residue numbers 424 through 993; a polynucleotide comprising the sequence of SEQ ID NO:6537, from cnucleotide residue numbers 424 through 993, wherein the nucleotide residue at 521 is
T;
c a polynucleotide comprising the sequence of SEQ ID NO:6537, from 00 nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 578 is M
C;
a polynucleotide comprising the sequence of SEQ ID NO:6537, from O 10 nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 649 is
G;
a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 653 is
T;
a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 721 is
A;
a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 781 is
A;
a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 788 is
G;
a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 989 is
A;
a polynucleotide comprising the sequence of SEQ ID NO:6533, from nucleotide residue numbers 423 through 707; and a polynucleotide comprising the sequence of SEQ ID NO:6545, from nucleotide residue numbers 83 through 427.
[0025B] The present provides a polynucleotide that encodes a protein according to the invention.
[0026] The invention provides polynucleotides corresponding or complementary to all or part of the PSCA genes, mRNAs, and/or coding sequences, preferably in isolated form, including polynucleotides encoding PSCA-related proteins and fragments of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 00
O
O
24, 25, or more than 25 contiguous amino acids; at least 30, 35, 40, 45, 50, 55, 60, 80, 85, 90, 95, 100 or more than 100 contiguous amino acids of a PSCA-related protein, as well as the peptides/proteins themselves; DNA, RNA, DNA/RNA hybrids, and related molecules, polynucleotides or oligonucleotides complementary or having at least a 90% homology to the PSCA genes or mRNA sequences or parts thereof, and c polynucleotides or oligonucleotides that hybridize to the PSCA genes, mRNAs, or to 00 PSCA-encoding polynucleotides. Also provided are means for isolating cDNAs and M the genes encoding PSCA. Recombinant DNA molecules containing PSCA polynucleotides, cells transformed or transduced with such molecules, and host-vector O 10 systems for the expression of PSCA gene products are also provided. The invention C further provides antibodies that bind to PSCA proteins and polypeptide fragments thereof, including polyclonal and monoclonal antibodies, murine and other mammalian antibodies, chimeric antibodies, humanized and fully human antibodies, and antibodies labeled with a detectable marker or therapeutic agent. In certain embodiments, there is a proviso that the entire nucleic acid sequence of Figure 2 is not encoded and/or the entire amino acid sequence of Figure 2 is not prepared. In certain embodiments, the entire nucleic acid sequence of Figure 2 is encoded and/or the entire amino acid sequence of Figure 2 is prepared, either of which are in respective human unit dose forms.
[0026A] The present invention further provides a recombinant expression vector comprising a polynucleotide of the invention.
[0026B] The present invention further provides a host cell that contains an expression vector of the invention.
[0026C] The present invention further provides a process for producing a PSCA protein comprising culturing a host cell of the invention under conditions sufficient for the production of the PSCA protein.
[0026D] The present invention further provides an isolated PSCA protein, wherein the PSCA protein comprises SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546.
[0026E] The present invention further provides an antibody or fragment thereof that immunospecifically binds to an epitope on a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546).
[0027] The invention further provides methods for detecting the presence and status of PSCA polynucleotides and proteins in various biological samples, as well as methods for identifying cells that express PSCA. A typical embodiment of this invention provides methods for monitoring PSCA gene products in a tissue or 00 c hematology sample having or suspected of having some form of growth dysregulation such as cancer.
[0027A] The present invention further provides a method for detecting, in a sample, the presence of a PSCA-related protein or a PSCA-related polynucleotide, comprising steps of: c contacting the sample with a substance that specifically binds to the PSCA- 00 related protein or to the PSCA-related polynucleotide, respectively; and, M determining that there is a complex of the substance with the PSCA-related CN protein or the substance with the PSCA-related polynucleotide, respectively.
O 10 [0027B] The present invention further provides a method of delivering a cytotoxic agent or a diagnostic agent to a cell that expresses a protein of Figure 2, said method comprising: providing the cytotoxic agent or the diagnostic agent conjugated to an antibody or fragment thereof according to the invention; and, exposing the cell to the antibody-agent or fragment-agent conjugate.
[0027C] The present invention further provides an in vitro method of delivering a cytotoxic agent to a cell expressing a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546) comprising providing an effective amount of an antibody according to the invention to the cell.
[0028] The invention further provides various immunogenic or therapeutic compositions and strategies for treating cancers that express PSCA such as cancers of tissues listed in Table 1, including therapies aimed at inhibiting the transcription, translation, processing or function of PSCA as well as cancer vaccines. In one aspect, the invention provides compositions, and methods comprising them, for treating a cancer that expresses PSCA in a human subject wherein the composition comprises a carrier suitable for human use and a human unit dose of one or more than one agent that inhibits the production or function of PSCA. Preferably, the carrier is a uniquely human carrier. In another aspect of the invention, the agent is a moiety that is immunoreactive with PSCA protein. Non-limiting examples of such moieties include, but are not limited to, antibodies (such as single chain, monoclonal, polyclonal, humanized, chimeric, or human antibodies), functional equivalents thereof (whether naturally occurring or synthetic), and combinations thereof. The antibodies can be conjugated to a diagnostic or therapeutic moiety. In another aspect, the agent is a small molecule as defined herein.
[0028A] The present invention further provides a composition comprising a pharmaceutically acceptable carrier and a protein of the invention.
00
O
O
10028B] The present invention further provides an in vitro method for detecting the presence of a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546) or polynucleotide (SEQ ID NO: 6533, SEQ ID SNO:6545, or SEQ ID NO:6537, wherein C521T, A578C, C649G, C653T, G721A, G781A, T788G, or G989A) in a test sample comprising: Cc contacting the sample with an antibody or polynucleotide, respectively, that OO specifically binds to the PSCA protein or polynucleotide, respectively; and
M
r detecting binding of PSCA protein or polynucleotide, respectively, in the sample thereto.
O 10 [0028C] The present invention further provides a method of generating a C mammalian immune response directed to a protein of Figure 2, the method comprising: exposing cells of the mammal's immune system to a portion of a) a PSCA-related protein and/or b) a nucleotide sequence that encodes said protein, whereby an immune response is generated to said protein.
[0028D] The present invention further provides a method of inducing an immune response to a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546), said method comprising: providing a PSCA protein epitope; contacting the epitope with an immune system T cell or B cell, whereby the immune system T cell or B cell is induced.
[0028E] The present invention further provides a composition comprising: a substance that a) modulates the status of a protein of Figure 2, or b) a molecule that is modulated by a protein of Figure 2, whereby the status of a cell that expresses a protein of Figure 2 is modulated.
10028F] The present invention further provides an in vitro method of inhibiting growth of a cell expressing a PSCA protein (SEQ ID NO: 6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546), comprising providing an effective amount of an antibody according to any one of claims 15, 16, or 18 to 25 to the cell, whereby the growth of the cell is inhibited.
[0028G] The present invention further provides an antibody or fragment thereof that specifically binds to a protein of Figure 2.
[0028H] The present invention further provides an antibody according to the invention, which is a human antibody, a humanized antibody or a chimeric antibody.
00 [00281] The present invention further provides a non-human transgenic animal Sthat produces an antibody according to the invention.
10028J] The present invention further provides a hybridoma that produces an Santibody according to the invention.
[0028K] The present invention further provides a method of inhibiting growth Cc of cancer cells that express a protein of Figure 2, the method comprising: 00 administering to the cells the composition according to the invention.
M [00291 In another aspect, the agent comprises one or more than one peptide Swhich comprises a cytotoxic T lymphocyte (CTL) epitope that binds an HLA class I O 10 molecule in a human to elicit a CTL response to PSCA and/or one or more than one C peptide which comprises a helper T lymphocyte (HTL) epitope which binds an HLA class II molecule in a human to elicit an HTL response. The peptides of the invention may be on the same or on one or more separate polypeptide molecules. In a further aspect of the invention, the agent comprises one or more than one nucleic acid molecule that expresses one or more than one of the CTL or HTL response stimulating peptides as described above. In yet another aspect of the invention, the one or more than one nucleic acid molecule may express a moiety that is immunologically reactive with PSCA as described above. The one or more than one nucleic acid molecule may also be, or encodes, a molecule that inhibits production of PSCA. Non-limiting examples of such molecules include, but are not limited to, those complementary to a nucleotide sequence essential for production of PSCA antisense sequences or molecules that form a triple helix with a nucleotide double helix essential for PSCA production) or a ribozyme effective to lyse PSCA mRNA.
[0029A] The present invention further provides a composition that comprises, consists essentially of, or consists of: a) a peptide of eight, nine, ten, or eleven contiguous amino acids of a protein of Figure 2; b) a peptide of Tables VIII-XXI; c) a peptide of Tables XXII to XLV; or, d) a peptide of Tables XLVI to XLIX.
10029B] The present invention further provides a composition comprising a polynucleotide that is fully complementary to a polynucleotide according to the invention.
[0029C] The present invention further provides use of a PSCA-related protein that comprises at least one T cell or at least one B cell epitope in the manufacture of a medicament for generating an immune response.
00 [0029D] The present invention further provides use of a PSCA protein (SEQ ID SNO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546) epitope for the preparation of a medicament to induce an immune response in a subject.
CI [0029E] The present invention further provides use of an antibody for the preparation of a medicament which delivers an agent to a cell expressing a PSCA cm protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 00 6546), wherein the antibody comprises an antibody according to the invention.
[0029F] The present invention further provides use of an effective amount of an antibody according to the invention for the preparation of a medicament which inhibits growth of a cell expressing a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, (N 6550, 6551, 6552, 6553, 6554, 6536, or 6546).
[0030] Note that to determine the starting position of any peptide set forth in Tables VIII-XXI and XXII to XLIX (collectively HLA Peptide Tables) respective to its parental protein, variant 1, variant 2, etc., reference is made to three factors: the particular variant, the length of the peptide in an HLA Peptide Table, and the Search Peptides in Table VII. Generally, a unique Search Peptide is used to obtain HLA peptides of a particular for a particular variant. The position of each Search Peptide relative to its respective parent molecule is listed in Table VII. Accordingly, if a Search Peptide begins at position one must add the value to each position in Tables VIII-XXI and XXII to XLIX to obtain the actual position of the HLA peptides in their parental molecule. For example, if a particular Search Peptide begins at position 150 of its parental molecule, one must add 150-1, 149 to each HLA peptide amino acid position to calculate the position of that amino acid in the parent molecule.
[0031] One embodiment of the invention comprises an HLA peptide, that occurs at least twice in Tables VIII-XXI and XXII to XLIX collectively, or an oligonucleotide that encodes the HLA peptide. Another embodiment of the invention comprises an HLA peptide that occurs at least once in Tables VIII-XXI and at least once in tables XXII to XLIX, or an oligonucleotide that encodes the HLA peptide.
[00321 Another embodiment of the invention is antibody epitopes, which comprise a peptide regions, or an oligonucleotide encoding the peptide region, that has one two, three, four, or five of the following characteristics: i) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that 00 includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.9, or having a value equal to 1.0, in the Hydrophilicity profile of Figure ii) a peptide region of at least 5 amino acids of a particular peptide of Figure S3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or less than 0.5, 0.4, 0.3, 0.2, c 0.1, or having a value equal to 0.0, in the Hydropathicity profile of Figure 6; 00 iii) a peptide region of at least 5 amino acids of a particular peptide of Figure M 3, in any whole number increment up to the full length of that protein in Figure 3, that
IND
includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, O 10 0.8, 0.9, or having a value equal to 1.0, in the Percent Accessible Residues profile of Figure 7; iv) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Average Flexibility profile of Figure 8; or v) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Beta-turn profile of Figure 9.
[0032A] Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application.
[0032B] Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
[0032C] Throughout this specification, the term "consisting essentially of' is intended to exclude elements which would materially affect the properties of the claimed composition.
Brief Description of the Drawings [0033] Figure 1. Intentionally Omitted.
[00341 Figure 2.
00 A) The cDNA and amino acid sequence of PSCA variant 1 (also called "PSCA v.1" or "PSCA variant is shown in Figure 2A. The start methionine is underlined.
The open reading frame extends from nucleic acid 18-389 including the stop codon.
SB) The cDNA and amino acid sequence of PSCA variant 2 (also called "PSCA is shown in Figure 2B. The codon for the start methionine is underlined. The c open reading frame extends from nucleic acid 56-427 including the stop codon.
00 C) The cDNA and amino acid sequence of PSCA variant 3 (also called "PSCA M is shown in Figure 2C. The codon for the start methionine is underlined. The Sopen reading frame extends from nucleic acid 423-707 including the stop codon.
O 10 D) The cDNA and amino acid sequence of PSCA variant 4 (also called "PSCA C is shown in Figure 2D. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 424-993 including the stop codon.
E) The cDNA and amino acid sequence of PSCA variant 5 (also called "PSCA is shown in Figure 2E. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 910-1479 including the stop codon.
F) The cDNA and amino acid sequence of PSCA variant 6 (also called "PSCA is shown in Figure 2F. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 83-427 including the stop codon.
G) SNP variants of PSCA v.2, PSCA v.7 through v.18. The PSCA v.7 through v.18 proteins have 123 amino acids. Variants PSCA v.7 through v.18 are variants with single nucleotide difference from PSCA v.2, and code for the same WO 2005/014780 PCT/US2004/017231 protein as v.2. Though these SNP variants are shown separately, they can also occur in any combinations and in any of the transcript variants listed above in Figures 2A through 2F.
H) SNP variants of PSCA v.4, PSCA v.19 through v.30. The PSCA v.19 through v.30 proteins have 189 amino acids. Variants PSCA v.19 through v.30 are variants with single nucleotide difference from PSCA v.4. PSCA v.9, v.11, v.24 and v.25 proteins differ from PSCA v.1 by one amino acid. PSCA v.23, v.28, v.29 and v.30 code for the same protein as v.4. Though these SNP variants are shown separately, they can also occur in any combinations and in any of the transcript variants v.3 and v.4.
[0035] Figure 3.
A) The amino acid sequence of PSCA v.1 is shown in Figure 3A; it has 123 amino acids.
B) The amino acid sequence of PSCA v.3 is shown in Figure 3B; it has 94 amino acids.
C) The amino acid sequence of PSCA v.4 is shown in Figure 3C; it has 189 amino acids.
D) The amino acid sequence of PSCA v.6 is shown in Figure 3D; it has 114 amino acids.
E) The amino acid sequence of PSCA v.19 is shown in Figure 3E; it has 189 amino acids.
F) The amino acid sequence of PSCA v.20 is shown in Figure 3F; it has 189 amino acids.
G) The amino acid sequence of PSCA v.21 is shown in Figure 3G; it has 189 amino acids.
H) The amino acid sequence of PSCA v.22 is shown in Figure 3H; it has 189 amino acids.
I) The amino acid sequence of PSCA v.24 is shown in Figure 31; it has 189 amino acids.
J) The amino acid sequence of PSCA v.25 is shown in Figure 3J; it has 189 amino acids.
K) The amino acid sequence of PSCA v.26 is shown in Figure 3k; it has 189 amino acids.
L) The amino acid sequence of PSCA v.27 is shown in Figure 3L; it has 189 amino acids.
[0036] As used herein, a reference to PSCA includes all variants thereof, including those shown in Figures 2, 3, 11, and 12 unless the context clearly indicates otherwise.
[0037] Figure 4. Alignment of PSCA v.4 with human Prostate Stem Cell Antigen (gi 27482160).
[0038] Figure 5. Figures Hydrophilicity amino acid profile of PSCAv.1, v.3, and v.4 determined by computer algorithm sequence analysis using the method ofHopp and Woods (Hopp Woods 1981. Proc. Natl. Acad. Sci.
U.S.A. 78:3824-3828) accessed on the Protscale website located on the World Wide Web at (expasy.ch/cgibin/protscale.pl) through the ExPasy molecular biology server.
[0039] Figure 6. Figures Hydropathicity amino acid profile of PSCAv.1, v.3, and v.4 determined by computer algorithm sequence analysis using the method of Kyte and Doolittle (Kyte Doclittle 1982. J. Mol. Biol.
157:105-132) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
[0040] Figure 7. Figures Percent accessible residues amino acid profile of PSCAv.1, v.3, and v.4 determined by computer algorithm sequence analysis using the method of Janin (Janin 1979 Nature 277:491-492) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
[0041] Figure 8. Figures Average flexibility amino acid profile of PSCAv.1, v.3, and v.4 determined by computer algorithm sequence analysis using the method of Bhaskaran and Ponnuswamy (Bhaskaran and Ponnuswamy 1988. Int. J. Pept. Protein Res. 32:242-255) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
WO 2005/014780 PCT/US2004/017231 [0042] Figure 9. Figures Beta-tum amino acid profile of PSCAv.1, v.3, and v.4 determined by computer algorithm sequence analysis using the method of Deleage and Roux (Deleage, Roux B. 1987 Protein Engineering 1:289-294) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
[0043] Figure 10. Exon compositions of transcript variants of PSCA. Variant PSCA v.2, v.3, v.4 and v.5 are transcript variants of v.1. Variant v.2 started transcription 47 bp further to the 5' end than v.1. Variant v.3 had a shorter exon 2 as compared to v.2. Variants v.4 and v.5 had an alternative first exon. Variant 5 kept the second intron as compared to v.4. The order of the potential exons on the human genome is shown at the bottom. Poly A tails were not shown in the figure. Ends of exons are shown above the boxes. Numbers in underneath the boxes correspond to those of PSCA v.2.
Lengths of introns and exons are not proportional.
[0044] Figure 11. Figure 11(a): Schematic alignment of protein variants of PSCA. Protein variants correspond to nucleotide variants. Nucleotide variants PSCAv.2, v.7 through v.18 coded the same protein as v.1. Variant v.5 coded the same protein as v.4 and protein v.3 was part of v.4. Nucleotide variants PSCA v.2, v.3, v.4 and v.5 were transcript variants of v.1, as shown in Figure 10. The SNP in v.2 that did not cause codon change in v.2 caused a codon change in v.3, v.4, and v.5. Single amino acid differences were indicated above the boxes. Black boxes represent the same sequence as PSCA v.1. Numbers underneath the box correspond to PSCAv.1. Figure 11(b): Schematic alignment of protein variants translated from SNP variants of PSCA v.4. Protein variants correspond to nucleotide variants. Nucleotide variants PSCA v.23, v.28, v.29 and v.30 coded the same protein as v.4. SNP in v.4 that resulted in an amino acid change in v.4 and also resulted in an amino acid change in v.5 and if occurring between aa 96-189, also in v.3. Single amino acid differences were indicated above the boxes. Black boxes represent the same sequence as PSCA v.4. Numbers underneath the box correspond to PSCA v.4.
[0045] Figure 12. Figure 12(a): Schematic alignment of SNP variants of PSCA v,2. Variants PSCA v.6 through v.18 are variants with single nucleotide differences as compared to variant v.2. Variant v.6 changed the ORF from 56-427 to 83-427. Though these SNP variants were shown separately, they could also occur in any combinations and in any transcript variants, such as v.4 shown in Fig. 12, that contained the base pairs. Numbers correspond to those of PSCA v.2.
Black box shows the same sequence as PSCA v.2. SNPs are indicated above the box. Figure 12(b): Schematic alignment of SNP variants of PSCA v.4. Variants PSCA v.19 through v.30 are variants with single nucleotide differences as compared to variant v.4 (ORF:424-993). Though these SNP variants were shown separately, they could also occur in any combinations and in any transcript variants that contained the base pairs, such as v.5 shown in Fig. 10,. Numbers correspond to those of PSCA v.4. Black box shows the same sequence as PSCA v.4. SNPs are indicated above the box.
[0046] Figure 13. Secondary structure and transmembrane domains prediction for PSCA protein variants.
Figures 13A, 13B, 13C, and 130: The secondary structure of PSCA protein variant 1 (SEQ ID NO:6532), variant 3 (SEQ ID NO:6536), variant 4 (SEQ ID NO:6540), and variant 6 (SEQ ID NO:6546) (Figures A-D, respectively) were predicted using the HNN Hierarchical Neural Network method (NPS@: Network Protein Sequence Analysis TIBS 2000 March Vol.
No 3 [291]:147-150 Combet Blanchet Geourjon C. and Deleage http://pbil.ibcp.fr/cgibininpsa_automat.pl?page=npsa nn.html), accessed from the ExPasy molecular biology server located on the World Wide Web at (.expasy.chftools/). This method predicts the presence and location of alpha helices, extended strands, and random coils from the primary protein sequence. The percent of each protein in a given secondary structure is also listed.
Figures 13E, 13G, 131, and 13K: Schematic representation of the probability of existence of transmembrane regions of PSCA variants 1,3,4, and 6, respectively based on the TMpred algorithm of Hofmann and Stoffel which utilizes TMBASE 8 WO 2005/014780 PCT/US2004/017231 Hofmann, W. Stoffel. TMBASE A database of membrane spanning protein segments Biol. Chem. Hoppe-Seyler 374:166, 1993). Figures 13F, 13H, 13J, and 13L: Schematic representation of the probability of the existence of transmembrane regions of PSCA variant 1, based on the TMHMM algorithm of Sonnhammer, von Heijne, and Krogh (Erik L.L. Sonnhammer, Gunnar von Heijne, and Anders Krogh: A hidden Markov model for predicting transmembrane helices in protein sequences. In Proc. of Sixth Int. Conf. on Intelligent Systems for Molecular Biology, p 175-182 Ed J. Glasgow, T.
Littlejohn, F. Major, R. Lathrop, D. Sankoff, and C. Sensen Menlo Park, CA: AAAI Press, 1998). The TMpred and TMHMM algorithms are accessed from the ExPasy molecular biology server (http://www.expasy.ch/tools/).
[0047] Figure 14. Expression of PSCA variants. Figure 14(A): Primers were designed to differentiate between PSCA v.1/v.2/v.4, PSCA v.3 and PSCA v.5. PSCA v.1/v.2/v.4 lead to a PCR product of 425 bp, PSCA v.3 leads to a PCR product of 300 bp, whereas PSCA v.5 leads to a PCR product of 910 bp in size. Figure 14(B): First strand cDNA was prepared from normal bladder, brain, heart, kidney, liver, lung, prostate, spleen, skeletal muscle, testis, pancreas, colon, stomach, pools of prostate cancer, bladder cancer, kidney cancer, colon cancer, lung cancer, ovary cancer, breast cancer, cancer metastasis, and pancreas cancer. Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using the variant specific primers was performed at 30 cycles of amplification. Results show expression of PSCA mainly in breast cancer, cancer metastasis, and pancreas cancer, and at lower level in colon cancer and lung cancer.
PSCA v.1/v.2/v.4 PCR product was detected in prostate cancer, bladder cancer, kidney cancer, colon cancer, lung cancer, ovary cancer, breast cancer, cancer metastasis, and pancreas cancer. Amongst normal tissues, PSCA v.1/v.2/v.4 PCR product was detected only in prostate, stomach and at lower level in kidney and lung, whereas PSCA v. 5 was not detected in any normal tissue. PSCA v.3 PCR detected product was not detected in any of the samples tested.
[0048] Figure 15. Expression of PSCA v.4 and PSCA v.5. Figure 15(A): Primers were designed to differentiate between PSCA v.4 and PSCA v.5. PSCA v.4 lead to a PCR product of 460 bp, whereas PSCA v.5 leads to a PCR product of 945 bp in size. Figure 15(B): First strand cDNA was prepared from normal bladder, brain, heart, kidney, liver, lung, prostate, spleen, skel. muscle, testis, pancreas, colon, stomach, pools of prostate cancer, bladder cancer, and multixenograft pool (prostate cancer, kidney cancer and bladder cancer xenografts). Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using the variant specific primers was performed at 30 cycles of amplification. Results show expression of PSCA v.4 in prostate cancer, bladder cancer, and multi-xenograft pool, normal kidney and prostate. PSCA v.5 was detected only in normal prostate and bladder cancer.
Modes of Carrying Out the Invention Outline of Sections Definitions II.) PSCA Polynucleotides II.A.) Uses of PSCA Polynucleotides II.A.1.) Monitoring of Genetic Abnormalities ll.A.2.) Antisense Embodiments II.A.3.) Primers and Primer Pairs II.A.4.) Isolation of PSCA-Encoding Nucleic Acid Molecules Recombinant Nucleic Acid Molecules and Host-Vector Systems III.) PSCA-related Proteins WO 2005/014780 PCT/US2004/017231 III.A.) Motif-bearing Protein Embodiments III.B.) Expression of PSCA-related Proteins III.C.) Modifications of PSCA-related Proteins III.D.) Uses of PSCA-related Proteins IV.) PSCA Antibodies PSCA Cellular Immune Responses VL) PSCA Transgenic Animals VII.) Methods for the Detection of PSCA ViII.) Methods for Monitoring the Status of PSCA-related Genes and Their Products IX.) Identification of Molecules That Interact With PSCA Therapeutic Methods and Compositions Anti-Cancer Vaccines PSCA as a Target for Antibody-Based Therapy PSCA as a Target for Cellular Immune Responses X.C.1. Minigene Vaccines X.C.2. Combinations of CTL Peptides with Helper Peptides X.C.3. Combinations of CTL Peptides with T Cell Priming Agents X.C.4. Vaccine Compositions Comprising DC Pulsed with CTL andlor HTL Peptides Adoptive Immunotherapy Administration of Vaccines for Therapeutic or Prophylactic Purposes XI.) Diagnostic and Prognostic Embodiments of PSCA.
XII.) Inhibition of PSCA Protein Function XII.A.) Inhibition of PSCA With Intracellular Antibodies XII.B.) Inhibition of PSCA with Recombinant Proteins XII.C.) Inhibition of PSCA Transcription or Translation XII.D.) General Considerations for Therapeutic Strategies XIII.) Identification, Characterization and Use of Modulators of PSCA XIV.) KITSIArticles of Manufacture Definitions: [0049] Unless otherwise defined, all terms of art, notations and other scientific terms or terminology used herein are intended to have the meanings commonly understood by those of skill in the art to which this invention pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art. Many of the techniques and procedures described or referenced herein are well understood and commonly employed using conventional methodology by those skilled in the art, such as, for example, the widely utilized molecular cloning methodologies described in Sambrook et al., Molecular Cloning: A Laboratory Manual 2nd.
edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. As appropriate, procedures involving the WO 2005/014780 PCT/US2004/017231 use of commercially available kits and reagents are generally carried out in accordance with manufacturer defined protocols and/or parameters unless otherwise noted.
[0050] The terms "advanced prostate cancer", "locally advanced prostate cancer", "advanced disease" and "locally advanced disease" mean prostate cancers that have extended through the prostate capsule, and are meant to include stage C disease under the American Urological Association (AUA) system, stage C1 C2 disease under the Whitmore- Jewett system, and stage T3 T4 and N+ disease under the TNM (tumor, node, metastasis) system. In general, surgery is not recommended for patients with locally advanced disease, and these patients have substantially less favorable outcomes compared to patients having clinically localized (organ-confined) prostate cancer. Locally advanced disease is clinically identified by palpable evidence of induration beyond the lateral border of the prostate, or asymmetry or induration above the prostate base. Locally advanced prostate cancer is presently diagnosed pathologically following radical prostatectomy if the tumor invades or penetrates the prostatic capsule, extends into the surgical margin, or invades the seminal vesicles.
[0051] "Altering the native glycosylation pattern" is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence PSCA (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical andlor enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence PSCA. In addition, the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present.
[0052] The term "analog" refers to a molecule which is structurally similar or shares similar or corresponding attributes with another molecule a PSCA-related protein). For example, an analog of a PSCA protein can be specifically bound by an antibody or T cell that specifically binds to PSCA.
[0053] The term "antibody" is used in the broadest sense. Therefore, an "antibody" can be naturally occurring or manmade such as monoclonal antibodies produced by conventional hybridoma technology. Anti-PSCA antibodies comprise monoclonal and polyclonal antibodies as well as fragments containing the antigen-binding domain and/or one or more complementarity determining regions of these antibodies.
[0054] An "antibody fragment" is defined as at least a portion of the variable region of the immunoglobulin molecule that binds to its target, the antigen-binding region. In one embodiment it specifically covers single anti-PSCA antibodies and cones thereof (including agonist, antagonist and neutralizing antibodies) and anti-PSCA antibody compositions with polyepitopic specificity.
[0055] The term "codon optimized sequences" refers to nucleotide sequences that have been optimized for a particular host species by replacing any codons having a usage frequency of less than about 20%. Nucleotide sequences that have been optimized for expression in a given host species by elimination of spurious polyadenylation sequences, elimination of exon/intron splicing signals, elimination of transposon-like repeats andlor optimization of GC content in addition to codon optimization are referred to herein as an "expression enhanced sequences." [0056] A "combinatorial library" is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical "building blocks" such as reagents. For example, a linear combinatorial chemical library, such as a polypeptide mutein) library, is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length the number of amino acids in a polypeptide compound). Numerous chemical compounds are synthesized through such combinatorial mixing of chemical building blocks (Gallop et al., J. Med. Chem. 37(9): 1233-1251 (1994)).
WO 2005/014780 PCT/US2004/017231 [0057] Preparation and screening of combinatorial libraries is well known to those of skill in the art. Such combinatorial chemical libraries include, but are not limited to, peptide libraries (see, U.S. Patent No. 5,010,175, Furka, Pept. Prot. Res. 37:487-493 (1991), Houghton et al., Nature, 354:84-88 (1991)), peptoids (PCT Publication No WO 91/19735), encoded peptides (PCT Publication WO 93/20242), random bio- oligomers (PCT Publication WO 92/00091), benzodiazepines Pat. No. 5,288,514), diversomers such as hydantoins, benzodiazepines and dipeptides (Hobbs et al., Proc. Nat. Acad. Sci. USA 90:6909-6913 (1993)), vinylogous polypeptides (Hagihara et al., J. Amer. Chem. Soc.
114:6568 (1992)), nonpeptidal peptidomimetics with a Beta-D-Glucose scaffolding (Hirschmann et al., J. Amer. Chem. Soc.
114:9217-9218 (1992)), analogous organic syntheses of small compound libraries (Chen et al., J. Amer. Chem. Soc.
116:2661 (1994)), oligocarbarnates (Cho, etal., Science 261:1303 (1993)), and/or peptidyl phosphonates (Campbell et al., J. Org. Chem. 59:658 (1994)). See, generally, Gordon et al., J. Med. Chem. 37:1385 (1994), nucleic acid libraries (see, Stratagene, Corp.), peptide nucleic acid libraries (see, U.S. Patent 5,539,083), antibody libraries (see, e.g., Vaughn et al., Nature Biotechnology 14(3): 309-314 (1996), and PCTIUS96/10287), carbohydrate libraries (see, Liang et al., Science 274:1520-1522 (1996), and U.S. Patent No. 5,593,853), and small organic molecule libraries (see, e.g., benzodiazepines, Baum, C&EN, Jan 18, page 33 (1993); isoprenoids, U.S. Patent No. 5,569,588; thiazolidinones and metathiazanones, U.S. Patent No. 5,549,974; pyrrolidines, U.S. Patent Nos. 5,525,735 and 5,519,134; morpholino compounds, U.S. Patent No. 5,506, 337; benzodiazepines, U.S. Patent No. 5,288,514; and the like).
[0058] Devices for the preparation of combinatorial libraries are commercially available (see, 357 NIPS, 390 NIPS, Advanced Chem Tech, Louisville KY; Symphony, Rainin, Woburn, MA; 433A, Applied Biosystems, Foster City, CA; 9050, Plus, Millipore, Bedford, NIA). A number of well-known robotic systems have also been developed for solution phase chemistries. These systems include automated workstations such as the automated synthesis apparatus developed by Takeda Chemical Industries, LTD. (Osaka, Japan) and many robotic systems utilizing robotic arms (Zymate H, Zymark Corporation, Hopkinton, Mass.; Orca, Hewlett-Packard, Palo Alto, Calif.), which mimic the manual synthetic operations performed by a chemist. Any of the above devices are suitable for use with the present invention. The nature and implementation of modifications to these devices (if any) so that they can operate as discussed herein will be apparent to persons skilled in the relevant art. In addition, numerous combinatorial libraries are themselves commercially available (see, ComGenex, Princeton, NJ; Asinex, Moscow, RU; Tripos, Inc., St. Louis, MO; ChemStar, Ltd, Moscow, RU; 3D Pharmaceuticals, Exton, PA; Martek Biosciences, Columbia, MD; etc.).
[0059] The term "cytotoxic agent" refers to a substance that inhibits or prevents the expression activity of cells, function of cells and/or causes destruction of cells. The term is intended to include radioactive isotopes chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof. Examples of cytotoxic agents include, but are not limited to auristatins, auromycins, maytansinoids, yttrium, bismuth, ricin, ricin A-chain, combrestatin, duocarmycins, dolostatins, doxorubicin, daunorubicin, taxol, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin, diphtheria toxin, Pseudomonas exotoxin (PE) A, PE40, abrin, abrin A chain, modeccin A chain, alpha-sarcin, gelonin, mitogellin, retstrictocin, phenomycin, enomycin, curicin, crotin, calicheamicin, Sapaonaria officinalis inhibitor, and glucocorticoid and other chemotherapeutic agents, as well as radioisotopes such as At 21 1 1131, 1125, Y 9 0, Re 1 86 Re 1 88 Sm1 53 Bi 2 2 lo 21 3 p32 and radioactive isotopes of Lu including Lu 1 77 Antibodies may also be conjugated to an anti-cancer pro-drug activating enzyme capable of converting the pro-drug to its active form.
WO 2005/014780 PCT/US2004/017231 [0060] The "gene product" is sometimes referred to herein as a protein or mRNA. For example, a "gene product of the invention" is sometimes referred to herein as a "cancer amino acid sequence", "cancer protein", "protein of a cancer listed in Table a "cancer mRNA", "mRNA of a cancer listed in Table etc. In one embodiment, the cancer protein is encoded by a nucleic acid of Figure 2. The cancer protein can be a fragment, or alternatively, be the full-length protein to the fragment encoded by the nucleic acids of Figure 2. In one embodiment, a cancer amino acid sequence is used to determine sequence identity or similarity. In another embodiment, the sequences are naturally occurring allelic variants of a protein encoded by a nucleic acid of Figure 2. In another embodiment, the sequences are sequence variants as further described herein.
[0061] "High throughput screening" assays for the presence, absence, quantification, or other properties of particular nucleic acids or protein products are well known to those of skill in the art. Similarly, binding assays and reporter gene assays are similarly well known. Thus, U.S. Patent No. 5,559,410 discloses high throughput screening methods for proteins; U.S. Patent No. 5,585,639 discloses high throughput screening methods for nucleic acid binding in arrays); while U.S. Patent Nos. 5,576,220 and 5,541,061 disclose high throughput methods of screening for ligand/antibody binding.
[0062] In addition, high throughput screening systems are commercially available (see, Amersham Biosciences, Piscataway, NJ; Zymark Corp., Hopkinton, MA; Air Technical Industries, Mentor, OH; Beckman Instruments, Inc. Fullerton, CA; Precision Systems, Inc., Natick, MA; etc.). These systems typically automate entire procedures, including all sample and reagent pipetting, liquid dispensing, timed incubations, and final readings of the microplate in detector(s) appropriate for the assay. These configurable systems provide high throughput and rapid start up as well as a high degree of flexibility and customization. The manufacturers of such systems provide detailed protocols for various high throughput systems.
Thus, Zymark Corp. provides technical bulletins describing screening systems for detecting the modulation of gene transcription, ligand binding, and the like.
[0063] The term "homolog" refers to a molecule which exhibits homology to another molecule, by for example, having sequences of chemical residues that are the same or similar at corresponding positions.
[0064] "Human Leukocyte Antigen" or "HLA" is a human class I or class II Major Histocompatibility Complex (MHC) protein (see, Stites, etal., IMMUNOLOGY, 8T ED., Lange Publishing, Los Altos, CA (1994).
[0065] The terms "hybridize", "hybridizing", "hybridizes" and the like, used in the context of polynucleotides, are meant to refer to conventional hybridization conditions, preferably such as hybridization in 50% formamide/6XSSC/0.1 SDS1100 pg/ml ssDNA, in which temperatures for hybridization are above 37 degrees C and temperatures for washing in 0.1XSSC/0.1% SDS are above 55 degrees C.
[0066] The phrases "isolated" or "biologically pure" refer to material which is substantially or essentially free from components which normally accompany the material as it is found in its native state. Thus, isolated peptides in accordance with the invention preferably do not contain materials normally associated with the peptides in their in situ environment. For example, a polynucleotide is said to be "isolated" when it is substantially separated from contaminant polynucleotides that correspond or are complementary to genes other than the PSCA genes or that encode polypeptides other than PSCA gene product or fragments thereof. A skilled artisan can readily employ nucleic acid isolation procedures to obtain an isolated PSCA polynucleotide. A protein is said to be "isolated," for example, when physical, mechanical or chemical methods are employed to remove the PSCA proteins from cellular constituents that are normally associated with the protein. A skilled artisan can readily employ standard purification methods to obtain an isolated PSCA protein. Alternatively, an isolated protein can be prepared by chemical means.
WO 2005/014780 PCT/LS2004/017231 [0067] The term "mammal" refers to any organism classified as a mammal, including mice, rats, rabbits, dogs, cats, cows, horses and humans, In one embodiment of the invention, the mammal is a mouse. In another embodiment of the invention, the mammal is a human.
[0068] The terms "metastatic prostate cancer" and "metastatic disease" mean prostate cancers that have spread to regional lymph nodes or to distant sites, and are meant to include stage D disease under the AUA system and stage TxNxM+ under the TNM system. As is the case with locally advanced prostate cancer, surgery is generally not indicated for patients with metastatic disease, and hormonal (androgen ablation) therapy is a preferred treatment modality. Patients with metastatic prostate cancer eventually develop an androgen-refractory state within 12 to 18 months of treatment initiation. Approximately half of these androgen-refractory patients die within 6 months after developing that status. The most common site for prostate cancer metastasis is bone. Prostate cancer bone metastases are often osteoblastic rather than osteolytic resulting in net bone formation). Bone metastases are found most frequently in the spine, followed by the femur, pelvis, rib cage, skull and humerus. Other common sites for metastasis include lymph nodes, lung, liver and brain. Metastatic prostate cancer is typically diagnosed by open or laparoscopic pelvic lymphadenectomy, whole body radionuclide scans, skeletal radiography, and/or bone lesion biopsy.
[0069] The term "modulator" or "test compound" or "drug candidate" or grammatical equivalents as used herein describe any molecule, protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for the capacity to directly or indirectly alter the cancer phenotype or the expression of a cancer sequence, a nucleic acid or protein sequences, or effects of cancer sequences signaling, gene expression, protein interaction, etc.) In one aspect, a modulator will neutralize the effect of a cancer protein of the invention. By "neutralize" is meant that an activity of a protein is inhibited or blocked, along with the consequent effect on the cell. In another aspect, a modulator will neutralize the effect of a gene, and its corresponding protein, of the invention by normalizing levels of said protein. In preferred embodiments, modulators alter expression profiles, or expression profile nucleic acids or proteins provided herein, or downstream effector pathways. In one embodiment, the modulator suppresses a cancer phenotype, e.g. to a normal tissue fingerprint. In another embodiment, a modulator induced a cancer phenotype. Generally, a plurality of assay mixtures is 'run in parallel with different agent concentrations to obtain a differential response to the various concentrations.
Typically, one of these concentrations serves as a negative control, at zero concentration or below the level of detection.
[0070] Modulators, drug candidates or test compounds encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 100 and less than about 2,500 Daltons. Preferred small molecules are less than 2000, or less than 1500 or less than 1000 or less than 500 D. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups. The candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Modulators also comprise biomolecules such as peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Particularly preferred are peptides. One class of modulators are peptides, for example of from about five to about 35 amino acids, with from about five to about 20 amino acids being preferred, and from about 7 to about being particularly preferred. Preferably, the cancer modulatory protein is soluble, includes a non-transmembrane region, and/or, has an N-terminal Cys to aid in solubility. In one embodiment, the C-terminus of the fragment is kept as a free acid and the N-terminus is a free amine to aid in coupling, to cysteine. In one embodiment, a cancer protein of the invention 14 WO 2005/014780 PCT/US2004/017231 is conjugated to an immunogenic agent as discussed herein. In one embodiment, the cancer protein is conjugated to BSA.
The peptides of the invention, of preferred lengths, can be linked to each other or to other amino acids to create a longer peptide/protein. The modulatory peptides can be digests of naturally occurring proteins as is outlined above, random peptides, or "biased" random peptides. In a preferred embodiment, peptidelprotein-based modulators are antibodies, and fragments thereof, as defined herein.
[0071] Modulators of cancer can also be nucleic acids. Nucleic acid modulating agents can be naturally occurring nucleic acids, random nucleic acids, or "biased" random nucleic acids. For example, digests of prokaryotic or eukaryotic genomes can be used in an approach analogous to that outlined above for proteins.
[0072] The term "monoclonal antibody" refers to an antibody obtained from a population of substantially homogeneous antibodies, the antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts.
[0073] A "motif', as in biological motif of a PSCA-related protein, refers to any pattern of amino acids forming part of the primary sequence of a protein, that is associated with a particular function protein-protein interaction, protein-DNA interaction, etc) or modification that is phosphorylated, glycosylated or amidated), or localization secretory sequence, nuclear localization sequence, etc.) or a sequence that is correlated with being immunogenic, either humorally or cellularly. A motif can be either contiguous or capable of being aligned to certain positions that are generally correlated with a certain function or property. In the context of HLA motifs, "motif" refers to the pattern of residues in a peptide of defined length, usually a peptide of from about 8 to about 13 amino acids for a class I HLA motif and from about 6 to about amino acids for a class II HLA motif, which is recognized by a particular HLA molecule. Peptide motifs for HLA binding are typically different for each protein encoded by each human HLA allele and differ in the pattern of the primary and secondary anchor residues.
[0074] A "pharmaceutical excipient" comprises a material such as an adjuvant, a carrier, pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservative, and the like.
[0075] "Pharmaceutically acceptable" refers to a non-toxic, inert, and/or composition that is physiologically compatible with humans or other mammals.
[0076] The term "polynucleotide" means a polymeric form of nucleotides of at least 10 bases or base pairs in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, and is meant to include single and double stranded forms of DNA and/or RNA. In the art, this term if often used interchangeably with "oligonucleotide". A polynucleotide can comprise a nucleotide sequence disclosed herein wherein thymidine as shown for example in Figure 2, can also be uracil this definition pertains to the differences between the chemical structures of DNA and RNA, in particular the observation that one of the four major bases in RNA is uracil instead of thymidine [0077] The term "polypeptide" means a polymer of at least about 4, 5, 6, 7, or 8 amino acids. Throughout the specification, standard three letter or single letter designations for amino acids are used. In the art, this term is often used interchangeably with "peptide" or "protein".
[0078] An HLA "primary anchor residue" is an amino acid at a specific position along a peptide sequence which is understood to provide a contact point between the immunogenic peptide and the HLA molecule. One to three, usually two, primary anchor residues within a peptide of defined length generally defines a "motif" for an immunogenic peptide. These residues are understood to fit in close contact with peptide binding groove of an HLA molecule, with their side chains buried in specific pockets of the binding groove. In one embodiment, for example, the primary anchor residues for an HLA class I molecule are located at position 2 (from the amino terminal position) and at the carboxyl terminal position of a 8, 9, 10, 11, WO 2005/014780 PCT/US2004/017231 or 12 residue peptide epitope in accordance with the invention. Alternatively, in another embodiment, the primary anchor residues of a peptide binds an HLA class II molecule are spaced relative to each other, rather than to the termini of a peptide, where the peptide is generally of at least 9 amino acids in length. The primary anchor positions for each motif and supermotif are set forth in Table IV. For example, analog peptides can be created by altering the presence or absence of particular residues in the primary and/or secondary anchor positions shown in Table IV. Such analogs are used to modulate the binding affinity and/or population coverage of a peptide comprising a particular HLA motif or supermotif.
[0079] "Radioisotopes" include, but are not limited to the following (non-limiting exemplary uses are also set forth): "Examples of Medical Isotopes: Isotope Description of use Actinium-225 Actinm225 See Thorium-229 (Th-229) (AC-225) Actinium-227 Parent of Radium-223 (Ra-223) which is an alpha emitter used to treat metastases in the skelet (AC-227) resulting from cancer breast and prostate cancers), and cancer radioimmunotherapy Bismuth-212 See Thorium-228 (Th-228) (Bi-212) Bismuth-213 See Thorium-229 (Th-229) (Bi-213) Cadmium-109 Cancer detection (Cd-109) Radiation source for radiotherapy of cancer, for food irradiators, and for sterilization of medical supplies Copper-64 A positron emitter used for cancer therapy and SPECT imaging (Cu-64) Copper-67 Beta/gamma emitter used in cancer radioimmunotherapy and diagnostic studies breast an (Cu-67) colon cancers, and lymphoma) Dysprosium-i 66 Dy- 166) Cancer radioimmunotherapy (Dy-166) Erbium-169 Rheumatoid arthritis treatment, particularly for the small joints associated with fingers and toes (Er-169) Europium-152 Radiation source for food irradiation and for sterilization of medical supplies (Eu-152) Europium-154 Radiation source for food irradiation and for sterilization of medical supplies (Eu-154) Gadolinim-153 Osteoporosis detection and nuclear medical quality assurance devices (Gd-153) GA-1 98) Implant and intracavity therapy of ovarian, prostate, and brain cancers (Au-198) Holmium-166 Multiple myeloma treatment in targeted skeletal therapy, cancer radioimmunotherapy, bone (Ho-166) marrow ablation, and rheumatoid arthritis treatment on d Osteoporosis detection, diagnostic imaging, tracer drugs, brain cancer treatment, radiolabeling, iodine-125 tumor imaging, mapping of receptors in the brain, interstitial radiation therapy, brachytherapy for (1-125) treatment of prostate cancer, determination of glomerular filtration rate (GFR), determination of plasma volume, detection of deep vein thrombosis of the legs odine-31 Thyroid function evaluation, thyroid disease detection, treatment of thyroid cancer as well as other oine non-malignant thyroid diseases Graves disease, goiters, and hyperthyroidism), treatment of (-131) leukemia, lymphoma, and other forms of cancer breast cancer) using radioimmunotherapy Iridium-192 Brachytherapy, brain and spinal cord tumor treatment, treatment of blocked arteries (lr-192) arteriosclerosis and restenosis), and implants for breast and prostate tumors WO 2005/014780 PCT/US2004/017231 Lutetium-177 Cancer radioimmunotherapy and treatment of blocked arteries arteriosclerosis and (Lu-177) restenosis) Parent of Technetium-99m (Tc-99m) which is used for imaging the brain, liver, lungs, heart, and Molybdenum-99 other organs. Currently, Tc-99m is the most widely used radioisotope used for diagnostic imaging (Mo-99) of various cancers and diseases involving the brain, heart, liver, lungs; also used in detection of deep vein thrombosis of the legs Osmium-1 94 T(0s-194) Cancer radioimmunotherapy Palladium-103 Paadi- 1 03 Prostate cancer treatment (Pd-103) Platinum-195m Patinum95m Studies on biodistribution and metabolism of cisplatin, a chemotherapeutic drug (Pt-1 95m) Polycythemia rubra vera (blood cell disease) and leukemia treatment, bone cancer Phosphorus-32 diagnosisltreatment; colon, pancreatic, and liver cancer treatment; radiolabeling nucleic acids for (P-32) in vitro research, diagnosis of superficial tumors, treatment of blocked arteries arteriosclerosis and restenosis), and intracavity therapy Phosphorus-33 Leukemia treatment, bone disease diagnosis/treatment, radiolabeling, and treatment of blocked (P-33) arteries arteriosclerosis and restenosis) Radium-223 (Ram-223 See Actinium-227 (Ac-227) (Ra-223) Rhenium-186 Bone cancer pain relief, rheumatoid arthritis treatment, and diagnosis and treatment of lymphoma (Re-186) and bone, breast, colon, and liver cancers using radioimmunotherapy Rhenium-188 Cancer diagnosis and treatment using radioimmunotherapy, bone cancer pain relief, treatment of (Re-188) rheumatoid arthritis, and treatment of prostate cancer Rhodium-105 (Rh-105) Cancer radioimmunotherapy Samarium-145 Smrm- 1 45 Ocular cancer treatment (Sm-1 45) Samarium- 53
(S
u m-153) Cancer radioimmunotherapy and bone cancer pain relief Scandium-47 (Sc-47) Cancer radioimmunotherapy and bone cancer pain relief Radiotracer used in brain studies, imaging of adrenal cortex by gamma-scintigraphy, lateral locations of steroid secreting tumors, pancreatic scanning, detection of hyperactive parathyroid glands, measure rate of bile acid loss from the endogenous pool Bone cancer detection and brain scans Strontium-89 (Stronm-89 Bone cancer pain relief, multiple myeloma treatment, and osteoblastic therapy (Sr-89) Techneim- 99 m See Molybdenum-99 (Mo-99) (To-99m) Thorium-228 Thorum-228 Parent of Bismuth-212 (Bi-212) which is an alpha emitter used in cancer radioimmunotherapy (Th-228) Thorium-229 Parent of Actinium-225 (Ac-225) and grandparent of Bismuth-213 (Bi-213) which are alpha (Th-229) emitters used in cancer radioimmunotherapy Thulium-170 Tuliu-170 Gamma source for blood irradiators, energy source for implanted medical devices
(T
m -170) Tin- 117m (Sn-117m) Cancer immunotherapy and bone cancer pain relief (Sn-1 17m) WO 2005/014780 PCT/US2004/017231 Tungsten-188 Parent for Rhenium-188 (Re-188) which is used for cancer diagnostics/treatment, bone cancer (W-188) pain relief, rheumatoid arthritis treatment, and treatment of blocked arteries arteriosclerosis and restenosis) Xenon-127 Neuroimaging of brain disorders, high resolution SPECT studies, pulmonary function tests, and (Xe-127) cerebral blood flow studies Ytterbium-175 Ytebum75 Cancer radioimmunotherapy (Yb-1 75) Microseeds obtained from irradiating Yttrium-89 (Y-89) for liver cancer treatment Yttrium-91 A gamma-emitting label for Yttrium-90 (Y-90) which is used for cancer radioimmunotherapy (Y-91) lymphoma, breast, colon, kidney, lung, ovarian, prostate, pancreatic, and inoperable liver cancers) [0080] By "randomized" or grammatical equivalents as herein applied to nucleic acids and proteins is meant that each nucleic acid and peptide consists of essentially random nucleotides and amino acids, respectively. These random peptides (or nucleic acids, discussed herein) can incorporate any nucleotide or amino acid at any position. The synthetic process can be designed to generate randomized proteins or nucleic acids, to allow the formation of all or most of the possible combinations over the length of the sequence, thus forming a library of randomized candidate bioactive proteinaceous agents.
[0081] In one embodiment, a library is "fully randomized," with no sequence preferences or constants at any position. In another embodiment, the library is a "biased random" library. That is, some positions within the sequence either are held constant, or are selected from a limited number of possibilities. For example, the nucleotides or amino acid residues are randomized within a defined class, of hydrophobic amino acids, hydrophilic residues, sterically biased (either small or large)'residues, towards the creation of nucleic acid binding domains, the creation of cysteines, for crosslinking, prolines for SH-3 domains, serines, threonines, tyrosines or histidines for phosphorylation sites, etc., or to purines, etc.
[0082] A "recombinant" DNA or RNA molecule is a DNA or RNA molecule that has been subjected to molecular manipulation in vitro.
[0083] Non-limiting examples of small molecules include compounds that bind or interact with PSCA, ligands including hormones, neuropeptides, chemokines, odorants, phospholipids, and functional equivalents thereof that bind and preferably inhibit PSCA protein function. Such non-limiting small molecules preferably have a molecular weight of less than about 10 kDa, more preferably below about 9, about 8, about 7, about 6, about 5 or about 4 kDa. In certain embodiments, small molecules physically associate with, or bind, PSCA protein; are not found in naturally occurring metabolic pathways; and/or are more soluble in aqueous than non-aqueous solutions [0084] "Stringency" of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured nucleic acid sequences to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
WO 2005/014780 PCT/US2004/017231 [0085] "Stringent conditions" or "high stringency conditions", as defined herein, are identified by, but not limited to, those that: employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 5000; employ during hybridization a denaturing agent, such as formamide, for example, 50% (vlv) formamide with 0.1% bovine serum albuminf0.1% Ficoll/0.1% mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42 oC; or employ formamide, 5 x SSC (0.75 M NaCI, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 |glml), 0.1% SDS, and 10% dextran sulfate at 42 OC, with washes at 42oC in 0.2 x SSC (sodium chloride/sodium. citrate) and 50% formamide at 55 oC, followed by a highstringency wash consisting of 0.1 x SSC containing EDTA at 55 oC. "Moderately stringent conditions" are described by, but not limited to, those in Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press, 1989, and include the use of washing solution and hybridization conditions temperature, ionic strength and %SDS) less stringent than those described above. An example of moderately stringent conditions is overnight incubation at 370C in a solution comprising: 20% formamide, 5 x SSC (150 mM NaCI, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 5 x Denhardt's solution, 10% dextran sulfate, and 20 mg/mL denatured sheared salmon sperm DNA, followed by washing the filters in 1 x SSC at about 37-500C, The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
[0086] An HLA "supermotif' is a peptide binding specificity shared by HLA molecules encoded by two or more HLA alleles. Overall phenotypic frequencies of HLA-supertypes in different ethnic populations are set forth in Table IV The non-limiting constituents of various supetypes are as follows: A2: A*0201, A*0202, A*0203, A*0204, A* 0205, A*0206, A*6802, A*6901, A*0207 A3: A3, All, A31, A*3301, A*6801, A*0301, A*1101, A*3101 B: B7, B*3501-03, B*51, B*5301, B*5401, B*5501, B*5502, B*5601, B*6701, B*7801, B*0702, B*5101, B*5602 B44: B*3701, B*4402, B*4403, B*60 (B*4001), B61 (B*4006) Al: A*0102, A*2604, A*3601, A*4301, A*8001 A24: A*24, A*30, A*2403, A*2404, A*3002, A*3003 B27: B*1401-02, B*1503, B*1509, B*1510, B*1518, B*3801-02, B*3901, B*3902, B*3903-04, B*4801-02, B*7301, B*2701-08 B58: B"1516, B*1517, B*5701, B*5702, B58 B62: B*4601, B52, B*1501 (B62), B*1502 (B75), B*1513 (B77) [0087] Calculated population coverage afforded by different HLA-supertype combinations are set forth in Table IV [0088] As used herein "to treat" or "therapeutic" and grammatically related terms, refer to any Improvement of any consequence of disease, such as prolonged survival, less morbidity, and/or a lessening of side effects which are the byproducts of an alternative therapeutic modality; full eradication of disease is not required.
[0089] A "transgenic animal" a mouse or rat) is an animal having cells that contain a transgene, which transgene was introduced into the animal or an ancestor of the animal at a prenatal, an embryonic stage. A "transgene" is a DNA that is integrated into the genome of a cell from which a transgenic animal develops.
[0090] As used herein, an HLA or cellular immune response "vaccine" is a composition that contains or encodes one or more peptides of the invention. There are numerous embodiments of such vaccines, such as a cocktail of one or WO 2005/014780 PCT/US2004/017231 more individual peptides; one or more peptides of the invention comprised by a polyepitopic peptide; or nucleic acids that encode such individual peptides or polypeptides, a minigene that encodes a polyepitopic peptide. The "one or more peptides" can include any whole unit integer from 1-150 or more, at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 16,17,18,19, 20, 21, 22, 23,24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41,42,43, 44,45,46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, or 150 or more peptides of the invention. The peptides or polypeptides can optionally be modified, such as by lipidation, addition of targeting or other sequences. HLA class I peptides of the invention can be admixed with, or linked to, HLA class II peptides, to facilitate activation of both cytotoxic T lymphocytes and helper T lymphocytes. HLA vaccines can also comprise peptide-pulsed antigen presenting cells, dendritic cells.
[0091] The term "variant" refers to a molecule that exhibits a variation from a described type or norm, such as a protein that has one or more different amino acid residues in the corresponding position(s) of a specifically described protein the PSCA protein shown in Figure 2 or Figure 3. An analog is an example of a variant protein. Splice isoforms and single nucleotides polymorphisms (SNPs) are further examples of variants.
[0092] The "PSCA-related proteins" of the invention include those specifically identified herein, as well as allelic variants, conservative substitution variants, analogs and homologs that can be isolated/generated and characterized without undue experimentation following the methods outlined herein or readily available in the art. Fusion proteins that combine parts of different PSCA proteins or fragments thereof, as well as fusion proteins of a PSCA protein and a heterologous polypeptide are also included. Such PSCA proteins are collectively referred to as the PSCA-related proteins, the proteins of the invention, or PSCA. The term "PSCA-related protein" refers to a polypeptide fragment or a PSCA protein sequence of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 amino acids; or, at least 30, 35, 40, 45, 50, 55, 60, 65, 85, 90, 95, 100, 105, 110, 115,120,125,130,135,140,145,150,155,160,165,170,175,180,185,190,195,200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, or 576 or more amino acids.
II.) PSCA Polynucleotides [0093] One aspect of the invention provides polynucleotides corresponding or complementary to all or part of a PSCA gene, mRNA, and/or coding sequence, preferably in isolated form, including polynucleotides encoding a PSCArelated protein and fragments thereof, DNA, RNA, DNA/RNA hybrid, and related molecules, polynucleotides or oligonucleotides complementary to a PSCA gene or mRNA sequence or a part thereof, and polynucleotides or oligonucleotides that hybridize to a PSCA gene, mRNA, or to a PSCA encoding polynucleotide (collectively, "PSCA polynucleotides"). In all instances when referred to in this section, T can also be U in Figure 2.
[0094] Embodiments of a PSCA polynucleotide include: a PSCA polynucleotide having the sequence shown in Figure 2, the nucleotide sequence of PSCA as shown in Figure 2 wherein T is U; at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in Figure 2; or, at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in Figure 2 where T is U. For example, embodiments of PSCA nucleotides comprise, without limitation: a polynucleotide comprising, consisting essentially of, or consisting of a sequence as shown in Figure 2, wherein T can also be U; (II) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown In Figure 2A, from nucleotide residue number 18 through nucleotide residue number 389, including the stop codon, wherein T can also be U; WO 2005/014780 PCT/US2004/017231 (Ill) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2B, from nucleotide residue number 56 through nucleotide residue number 427, including the stop codon, wherein T can also be U; (IV) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2C, from nucleotide residue number 423 through nucleotide residue number 707, including the a stop codon, wherein T can also be U; a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2D, from nucleotide residue number 424 through nucleotide residue number 993, including the stop codon, wherein T can also be U; (VI) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2E, from nucleotide residue number 910 through nucleotide residue number 1479, including the stop codon, wherein T can also be U; (VII) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2F, from nucleotide residue number 83 through nucleotide residue number 427, including the stop codon, wherein T can also be U; (VIII) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2G, from nucleotide residue number 56 through nucleotide residue number 427, including the stop codon, wherein T can also be U; (IX) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2H, from nucleotide residue number 424 through nucleotide residue number 993, including the stop codon, wherein T can also be U; a polynucleotide that encodes a PSCA-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% homologous to an entire amino acid sequence shown in Figure 2A-H; (XI) a polynucleotide that encodes a PSCA-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% identical to an entire amino acid sequence shown in Figure 2A-H; (XII) polynucleotide that encodes at least one peptide set forth in Tables VIIl-XXI and XXII-XLIX; (XIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figures 3A in any whole number increment up to 123 that includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14,15, 16,17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0,5 in the Hydrophilicity profile of Figure (XIV) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3A in any whole number increment up to 123 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16,17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; (XV) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3A in any whole number increment up to 123 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, WO 2005/014780 PCT/US2004/017231 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XVI) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12,13, 14, 15, 16,17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3A in any whole number increment up to 123 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9,10, 11, 12, 13,14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; (XVII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12,13,14,15, 16,17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3A in any whole number increment up to 123 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XVIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3B in any whole number increment up to 94 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,11, 12,13, 14, 15, 16,17, 18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of Figure (XIX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3B in any whole number increment up to 94 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12,13, 14,15, 16,17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; (XX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16,17, 18, 19, 20, 21,22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3B in any whole number increment up to 94 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12,13, 14,15, 16,17, 18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XXI) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30,31, 32, 33, 34, 35 amino acids of a peptide of Figure 3B in any whole number increment up to 94 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,11, 12,13, 14, 15, 16, 17, 18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; (XXII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3B in any whole number increment up to 94 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,11, 12, 13, 14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XXIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figures 3C, 3E-3L in any whole number increment up to 189 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of Figure (XXIV) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figures 3C, 3E-3L in any whole 22 WO 2005/014780 PCT/US2004/017231 number increment up to 189 that includes 1, 2,3, 4,5, 6,7,8,9,10,11,12,13,14,15,16,17,18,19,20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; (XXV) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figures 3C, 3E-3L in any whole number increment up to 189 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16,17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XXVI) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9,10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figures 3C, 3E-3L in any whole number increment up to 189 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14,15,16, 17,18,19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; (XXVII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figures 3C, 3E-3L in any whole number increment up to 189 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9,10, 11, 12,13,14, 15,16,17,18,19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XXVIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9,10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3D in any whole number increment up to 114 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,11, 12,13, 14,15, 16,17,18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of Figure (XXIX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3D in any whole number increment up to 114 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; (XXX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9,10, 11, 12, 13, 14, 15,16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3D in any whole number increment up to 114 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,11, 12,13, 14,15,16,17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XXXI) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3D in any whole number increment up to 114 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,11, 12, 13, 14, 15,16,17, 18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; (XXXII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9,10, 11, 12, 13,14, 15,16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of Figure 3D in any whole number 23 WO 2005/014780 PCT/US2004/017231 increment up to 114 that includes 1, 2, 3,4,5, 6, 7,8, 9,10,11,12,13,14,15,16,17, 18,19, 20, 21,22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XXXIII) a polynucleotide that is fully complementary to a polynucleotide of any one of (I)-(XXXII).
(XXXIV) a peptide that is encoded by any of to (XXXIII); and; (XXXV) a composition comprising a polynucleotide of any of (I)-(XXXIII) or peptide of (XXXIV) together with a pharmaceutical excipient and/or in a human unit dose form; (XXXVI) a method of using a polynucleotide of any (I)-(XXXIII) or peptide of (XXXIV) or a composition of (XXXV) in a method to modulate a cell expressing PSCA,; (XXXVII) a method of using a polynucleotide of any (I)-(XXXIII) or peptide of (XXXIV) or a composition of (XXXV) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing PSCA; (XXXVIII) a method of using a polynucleotide of any (I)-(XXXIII) or peptide of (XXXIV) or a composition of (XXXV) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing PSCA, said cell from a cancer of a tissue listed in Table I (XXXIX) a method of using a polynucleotide of any (I)-(XXXIII) or peptide of (XXXIV) or a composition of (XXXV) in a method to diagnose, prophylax, prognose, or treat a a cancer (XL) a method of using a polynucleotide of any (I)-(XXXIII) or peptide of (XXXIV) or a composition of (XXXV) in a method to diagnose, prophylax, prognose, or treat a a cancer of a tissue listed in Table I; and,; (XLI) a method of using a polynucleotide of any (I)-(XXXIII) or peptide of (XXXIV) or a composition of (XXXV) in a method to identify or characterize a modulator of a cell expressing PSCA.; [0095] As used herein, a range is understood to disclose specifically all whole unit positions thereof.
[0096] Typical embodiments of the invention disclosed herein include PSCA polynucleotides that encode specific portions of PSCA mRNA sequences (and those which are complementary to such sequences) such as those that encode the proteins and/or fragments thereof, for example: 4, 5, 6, 7, 8, 9,10, 11, 12, 13,14, 15,16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 80, 85, 90, 95, 100, 105, 110, 115, 120, and 123 or more contiguous amino acids of PSCA variant 1; the maximal lengths relevant for other variants are: variant 3, 94 amino acids; variant 4, 189 amino acids, variant 6, 114 amino acids, variant 19, 189 amino acids, variant 20, 189 amoni acids, variant 21,189 amino acids, variant 22, 189 amino acids, variant 24, 189 amino acids, variant 25, 189 amino acids, variant 26,189 amino acids, and variant 27, 189 amino acids.
[0097] For example, representative embodiments of the invention disclosed herein include: polynucleotides and their encoded peptides themselves encoding about amino acid 1 to about amino acid 10 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 10 to about amino acid 20 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 20 to about amino acid 30 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 30 to about amino acid 40 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 40 to about amino acid 50 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 50 to about amino acid 60 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 60 to about amino acid 70 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 70 to about amino acid 80 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 80 to about amino acid 90 of the PSCA protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 90 to about amino acid 100 of the PSCA protein shown in Figure 2 WO 2005/014780 PCT/US2004/017231 or Figure 3, in increments of about 10 amino acids, ending at the carboxyl terminal amino acid set forth in Figure 2 or Figure 3. Accordingly, polynucleotides encoding portions of the amino acid sequence (of about 10 amino acids), of amino acids, 100 through the carboxyl terminal amino acid of the PSCA protein are embodiments of the invention. Wherein it is understood that each particular amino acid position discloses that position plus or minus five amino acid residues.
[0098] Polynucleotides encoding relatively long portions of a PSCA protein are also within the scope of the invention.
For example, polynucleotides encoding from about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 30, or or 50 etc.) of the PSCA protein "or variant" shown in Figure 2 or Figure 3 can be generated by a variety of techniques well known in the art. These polynucleotide fragments can include any portion of the PSCA sequence as shown in Figure 2.
[0100] Additional illustrative embodiments of the invention disclosed herein include PSCA polynucleotide fragments encoding one or more of the biological motifs contained within a PSCA protein "or variant" sequence, including one or more of the motif-bearing subsequences of a PSCA protein "or variant" set forth in Tables VIII-XXI and XXII-XLIX. In another embodiment, typical polynucleotide fragments of the invention encode one or more of the regions of PSCA protein or variant that exhibit homology to a known molecule. In another embodiment of the invention, typical polynucleotide fragments can encode one or more of the PSCA protein or variant N-glycosylation sites, cAMP and cGMP-dependent protein kinase phosphorylation sites, casein kinase II phosphorylation sites or N-myristoylation site and amidation sites.
[0101] Note that to determine the starting position of any peptide set forth in Tables VIII-XXI and Tables XXII to XLIX (collectively HLA Peptide Tables) respective to its parental protein, variant 1, variant 2, etc., reference is made to three factors: the particular variant, the length of the peptide in an HLA Peptide Table, and the Search Peptides listed in Table VII. Generally, a unique Search Peptide is used to obtain HLA peptides for a particular variant. The position of each Search Peptide relative to its respective parent molecule is listed in Table VII. Accordingly, if a Search Peptide begins at position one must add the value "X minus 1" to each position in Tables VIII-XXI and Tables XXII-IL to obtain the actual position of the HLA peptides in their parental molecule. For example if a particular Search Peptide begins at position 150 of its parental molecule, one must add 150 1, 149 to each HLA peptide amino acid position to calculate the position of that amino acid in the parent molecule.
II.A.) Uses of PSCA Polynucleotides II.A.1.) Monitoring of Genetic Abnormalities [0102] The polynucleotides of the preceding paragraphs have a number of different specific uses. The human PSCA gene maps to the chromosomal location set forth in the Example entitled "Chromosomal Mapping of PSCA." For example, because the PSCA gene maps to this chromosome, polynucleotides that encode different regions of the PSCA proteins are used to characterize cytogenetic abnormalities of this chromosomal locale, such as abnormalities that are identified as being associated with various cancers. In certain genes, a variety of chromosomal abnormalities including rearrangements have been identified as frequent cytogenetic abnormalities in a number of different cancers (see e.g.
Krajinovic et Mutat. Res. 382(3-4): 81-83 (1998); Johansson et al, Blood 86(10): 3905-3914 (1995) and Finger et a., P.N.A.S. 85(23): 9158-9162 (1988)). Thus, polynucleotides encoding specific regions of the PSCA proteins provide new tools that can be used to delineate, with greater precision than previously possible, cytogenetic abnormalities in the chromosomal region that encodes PSCA that may contribute to the malignant phenotype. In this context, these polynucleotides satisfy a need in the art for expanding the sensitivity of chromosomal screening in order to identify more WO 2005/014780 PCT/US2004/017231 subtle and less common chromosomal abnormalities (see e.g. Evans et al, Am. J. Obstet. Gynecol 171(4): 1055-1057 (1994)).
[0103] Furthermore, as PSCA was shown to be highly expressed In prostate and other cancers, PSCA polynucleotides are used in methods assessing the status of PSCA gene products in normal versus cancerous tissues.
Typically, polynucleotides that encode specific regions of the PSCA proteins are used to assess the presence of perturbations (such as deletions, insertions, point mutations, or alterations resulting in a loss of an antigen etc.) in specific regions of the PSCA gene, such as regions containing one or more motifs. Exemplary assays include both RT-PCR assays as well as single-strand conformation polymorphism (SSCP) analysis (see, Marrogi et J. Cutan. Pathol. 26(8): 369- 378 (1999), both of which utilize polynucleotides encoding specific regions of a protein to examine these regions within the protein.
II.A.2.) Antisense Embodiments [0104] Other specifically contemplated nucleic acid related embodiments of the invention disclosed herein are genomic DNA, cDNAs, ribozymes, and antisense molecules, as well as nucleic acid molecules based on an alternative backbone, or including alternative bases, whether derived from natural sources or synthesized, and include molecules capable of inhibiting the RNA or protein expression of PSCA. For example, antisense molecules can be RNAs or other molecules, including peptide nucleic acids (PNAs) or non-nucleic acid molecules such as phosphorothioate derivatives that specifically bind DNA or RNA in a base pair-dependent manner. A skilled artisan can readily obtain these classes of nucleic acid molecules using the PSCA polynucleotides and polynucleotide sequences disclosed herein.
[0105] Antisense technology entails the administration of exogenous oligonucleotides that bind to a target polynucleotide located within the cells. The term "antisense" refers to the fact that such oligonucleotides are complementary to their intracellular targets, PSCA. See for example, Jack Cohen, Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press, 1989; and Synthesis 1:1-5 (1988). The PSCA antisense oligonucleotides of the present invention include derivatives such as S-oligonucleotides (phosphorothioate derivatives or S-oligos, see, Jack Cohen, supra), which exhibit enhanced cancer cell growth inhibitory action. S-oligos (nucleoside phosphorothioates) are isoelectronic analogs of an oligonucleotide (O-oligo) in which a nonbridging oxygen atom of the phosphate group is replaced by a sulfur atom. The S-oligos of the present invention can be prepared by treatment of the corresponding Ooligos with 3H-1,2-benzodithiol-3-one-1,1-dioxide, which is a sulfur transfer reagent. See, lyer, R. P. et al., J. Org.
Chem. 55:4693-4698 (1990); and lyer, R. P. et al., J. Am. Chem. Soc. 112:1253-1254 (1990). Additional PSCA antisense oligonucleotides of the present invention include morpholino antisense oligonucleotides known in the art (see, e.g., Partridge et aL, 1996, Antisense Nucleic Acid Drug Development 6: 169-175).
[0106] The PSCA antisense oligonucleotides of the present invention typically can be RNA or DNA that is complementary to and stably hybridizes with the first 100 5' codons or last 100 3' codons of a PSCA genomic sequence or the corresponding mRNA. Absolute complementarity is not required, although high degrees of complementarity are preferred. Use of an oligonucleotide complementary to this region allows for the selective hybridization to PSCA mRNA and not to mRNA specifying other regulatory subunits of protein kinase. In one embodiment, PSCA antisense oligonucleotides of the present invention are 15 to 30-mer fragments of the antisense DNA molecule that have a sequence that hybridizes to PSCA mRNA. Optionally, PSCA antisense oligonucleotide is a 30-mer oligonucleotide that is complementary to a region in the first 10 5' codons or last 10 3' codons of PSCA. Alternatively, the antisense molecules WO 2005/014780 PCT/US2004/017231 are modified to employ ribozymes in the inhibition of PSCA expression, see, L. A. Couture D. T. Stinchcomb; Trends Genet 12: 510-515 (1996).
II.A.3.) Primers and Primer Pairs [0107] Further specific embodiments of these nucleotides of the invention include primers and primer pairs, which allow the specific amplification of polynucleotides of the invention or of any specific parts thereof, and probes that selectively or specifically hybridize to nucleic acid molecules of the invention or to any part thereof. Probes can be labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme. Such probes and primers are used to detect the presence of a PSCA polynucleotide in a sample and as a means for detecting a cell expressing a PSCA protein.
[0108] Examples of such probes include polypeptides comprising all or part of the human PSCA cDNA sequence shown in Figure 2. Examples of primer pairs capable of specifically amplifying PSCA mRNAs are also described in the Examples. As will be understood by the skilled artisan, a great many different primers and probes can be prepared based on the sequences provided herein and used effectively to amplify and/or detect a PSCA mRNA.
[0109] The PSCA polynucleotides of the invention are useful for a variety of purposes, including but not limited to their use as probes and primers for the amplification and/or detection of the PSCA gene(s), mRNA(s), or fragments thereof; as reagents for the diagnosis and/or prognosis of prostate cancer and other cancers; as coding sequences capable of directing the expression of PSCA polypeptides; as tools for modulating or inhibiting the expression of the PSCA gene(s) and/or translation of the PSCA transcript(s); and as therapeutic agents.
[0110] The present invention includes the use of any probe as described herein to identify and isolate a PSCA or PSCA related nucleic acid sequence from a naturally occurring source, such as humans or other mammals, as well as the isolated nucleic acid sequence per se, which would comprise all or most of the sequences found in the probe used.
II.A.4.) Isolation of PSCA-Encoding Nucleic Acid Molecules [0111] The PSCA cDNA sequences described herein enable the isolation of other polynucleotides encoding PSCA gene product(s), as well as the isolation of polynucleotides encoding PSCA gene product homologs, alternatively spliced isoforms, allelic variants, and mutant forms of a PSCA gene product as well as polynucleotides that encode analogs of PSCA-related proteins. Various molecular cloning methods that can be employed to isolate full length cDNAs encoding a PSCA gene are well known (see, for example, Sambrook, J. et Molecular Cloning: A Laboratory Manual, 2d edition, Cold Spring Harbor Press, New York, 1989; Current Protocols in Molecular Biology. Ausubel et Eds., Wiley and Sons, 1995). For example, lambda phage cloning methodologies can be conveniently employed, using commercially available cloning systems Lambda ZAP Express, Stratagene). Phage clones containing PSCA gene cDNAs can be identified by probing with a labeled PSCA cDNA or a fragment thereof. For example, in one embodiment, a PSCA cDNA Figure 2) or a portion thereof can be synthesized and used as a probe to retrieve overlapping and full-length cDNAs corresponding to a PSCA gene. A PSCA gene itself can be isolated by screening genomic DNA libraries, bacterial artificial chromosome libraries (BACs), yeast artificial chromosome libraries (YACs), and the like, with PSCA DNA probes or primers.
WO 2005/014780 PCT/US2004/017231 Recombinant Nucleic Acid Molecules and Host-Vector Systems [0112] The invention also provides recombinant DNA or RNA molecules containing a PSCA polynucleotide, a fragment, analog or homologue thereof, including but not limited to phages, plasmids, phagemids, cosmids, YACs, BACs, as well as various viral and non-viral vectors well known in the art, and cells transformed or transfected with such recombinant DNA or RNA molecules. Methods for generating such molecules are well known (see, for example, Sambrook et 1989, supra).
[0113] The invention further provides a host-vector system comprising a recombinant DNA molecule containing a PSCA polynucleotide, fragment, analog or homologue thereof within a suitable prokaryotic or eukaryotic host cell.
Examples of suitable eukaryotic host cells include a yeast cell, a plant cell, or an animal cell, such as a mammalian cell or an insect cell a baculovirus-infectible cell such as an Sf9 or HighFive cell). Examples of suitable mammalian cells include various prostate cancer cell lines such as DU145 and TsuPrl, other transfectable or transducible prostate cancer cell lines, primary cells (PrEC), as well as a number of mammalian cells routinely used for the expression of recombinant proteins COS, CHO, 293, 293T cells). More particularly, a polynucleotide comprising the coding sequence of PSCA or a fragment, analog or homolog thereof can be used to generate PSCA proteins or fragments thereof using any number of host-vector systems routinely used and widely known in the art.
[0114] A wide range of host-vector systems suitable for the expression of PSCA proteins or fragments thereof are available, see for example, Sambrook et al., 1989, supra; Current Protocols in Molecular Biology, 1995, supra). Preferred vectors for mammalian expression include but are not limited to pcDNA 3.1 myc-His-tag (Invitrogen) and the retroviral vector pSRcdkneo (Muller et al., 1991, MCB 11:1785). Using these expression vectors, PSCA can be expressed in several prostate cancer and non-prostate cell lines, including for example 293, 293T, rat-1, NIH 3T3 and TsuPrI. The host-vector systems of the invention are useful for the production of a PSCA protein or fragment thereof. Such host-vector systems can be employed to study the functional properties of PSCA and PSCA mutations or analogs.
[0115] Recombinant human PSCA protein or an analog or homolog or fragment thereof can be produced by mammalian cells transfected with a construct encoding a PSCA-related nucleotide. For example, 293T cells can be transfected with an expression plasmid encoding PSCA or fragment, analog or homolog thereof, a PSCA-related protein is expressed in the 293T cells, and the recombinant PSCA protein is isolated using standard purification methods affinity purification using anti-PSCA antibodies). In another embodiment, a PSCA coding sequence is subcloned into the retroviral vector pSRaMSVtkneo and used to infect various mammalian cell lines, such as NIH 3T3, TsuPrl, 293 and rat-1 in order to establish PSCA expressing cell lines. Various other expression systems well known in the art can also be employed. Expression constructs encoding a leader peptide joined in frame to a PSCA coding sequence can be used for the generation of a secreted form of recombinant PSCA protein.
[0116] As discussed herein, redundancy in the genetic code permits variation in PSCA gene sequences. In particular, it is known in the art that specific host species often have specific codon preferences, and thus one can adapt the disclosed sequence as preferred for a desired host. For example, preferred analog codon sequences typically have rare codons codons having a usage frequency of less than about 20% in known sequences of the desired host) replaced with higher frequency codons. Codon preferences for a specific species are calculated, for example, by utilizing codon usage tables available on the INTERNET such as at URL dna.affrc.go.jp/~nakamura/codon.html.
[0117] Additional sequence modifications are known to enhance protein expression in a cellular host. These include elimination of sequences encoding spurious polyadenylation signals, exon/intron splice site signals, transposon-like WO 2005/014780 PCT/US2004/017231 repeats, and/or other such well-characterized sequences that are deleterious to gene expression. The GC content of the sequence is adjusted to levels average for a given cellular host, as calculated by reference to known genes expressed in the host cell. Where possible, the sequence is modified to avoid predicted hairpin secondary mRNA structures. Other useful modifications include the addition of a translational initiation consensus sequence at the start of the open reading frame, as described in Kozak, Mol. Cell Biol., 9:5073-5080 (1989). Skilled artisans understand that the general rule that eukaryotic ribosomes initiate translation exclusively at the 5' proximal AUG codon is abrogated only under rare conditions (see, Kozak PNAS 92(7): 2662-2666, (1995) and Kozak NAR 15(20): 8125-8148 (1987)).
II.) PSCA-related Proteins [0118] Another aspect of the present invention provides PSCA-related proteins. Specific embodiments of PSCA proteins comprise a polypeptide having all or part of the amino acid sequence of human PSCA as shown in Figure 2 or Figure 3. Alternatively, embodiments of PSCA proteins comprise variant, homolog or analog polypeptides that have alterations in the amino acid sequence of PSCA shown in Figure 2 or Figure 3.
[0119] Embodiments of a PSCA polypeptide include: a PSCA polypeptide having a sequence shown in Figure 2, a peptide sequence of a PSCA as shown in Figure 2 wherein T is U; at least 10 contiguous nucleotides of a polypeptide having the sequence as shown in Figure 2; or, at least 10 contiguous peptides of a polypeptide having the sequence as shown in Figure 2 where T is U. For example, embodiments of PSCA peptides comprise, without limitation:(l) a protein comprising, consisting essentially of, or consisting of an amino acid sequence as shown in Figure 2A-H or Figure 3A-L; (II) a PSCA-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% homologous to an entire amino acid sequence shown in Figure 2A-H or 3A-L; (III) a PSCA-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% identical to an entire amino acid sequence shown in Figure 2A-H or 3A-L; (IV) a protein that comprises at least one peptide set forth in Tables VIII to XLIX, optionally with a proviso that it is not an entire protein of Figure 2; a protein that comprises at least one peptide set forth in Tables VIII-XXI, collectively, which peptide is also set forth in Tables XXII to XLIX, collectively, optionally with a proviso that it is not an entire protein of Figure 2; (VI) a protein that comprises at least two peptides selected from the peptides set forth in Tables VIII-XLIX, optionally with a proviso that it is not an entire protein of Figure 2; (VII) a protein that comprises at least two peptides selected from the peptides set forth in Tables VIII to XLIX collectively, with a proviso that the protein is not a contiguous sequence from an amino acid sequence of Figure 2; (VIII) a protein that comprises at least one peptide selected from the peptides set forth in Tables VIII-XXI; and at least one peptide selected from the peptides set forth in Tables XXII to XLIX, with a proviso that the protein is not a contiguous sequence from an amino acid sequence of Figure 2; (IX) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16,17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3A in any whole number increment up to 123 respectively that includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of Figure WO 2005/014780 PCT/US2004/017231 a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12,13,14, 15,16,17,18, 19,20, 21,22, 23,24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3A, in any whole number increment up to 123 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; (XI) a polypeptide comprising at least 5, 6, 7, 8, 9,10, 11, 12,13,14, 15,16, 17,18,19, 20,21,22,23,24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3A, in any whole number increment up to 123 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14,15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XII) a polypeptide comprising atleast 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3A, in any whole number increment up to 123respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; (XIII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, amino acids of a protein of Figure 3A in any whole number increment up to 123 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XIV) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3B, in any whole number increment up to 94 respectively that includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of Figure (XV) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16,17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3B, in any whole number increment up to 94 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; (XVI) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12,13, 14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3B, in any whole number increment up to 94 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15,16, 17,18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XVII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12,13, 14,15, 16,17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3B, in any whole number increment up to 94 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; WO 2005/014780 PCT/LS2004/017231 (XVIII) a polypeptide comprising at least 5, 6, 7,8, 9, 10,11,12,13,14,15,16,17, 18,19, 20, 21, 22,23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, amino acids of a protein of Figure 3B in any whole number increment up to 94 respectively that includes at least at least 1,2, 3,4,5,6,7, 8,9,10, 11,12,13,14,15,16,17,18,19, 20, 21,22, 23,24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XIX) a polypeptide comprising at least 5, 6,7,8,9, 10,11, 12,13, 14,15,16,17,18,19,20,21, 22,23,24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3C, 3E-3L in any whole number increment up to 189 respectively that includes at least 1,2,3,4,5,6,7, 8, 9, 10,11,12,13, 14,15, 16,17, 18,19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of Figure (XX) a polypeptide comprising at least 5, 6, 7, 8, 9, 10,11, 12,13, 14,15,16,17,18,19, 20,21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3C, 3E-3L in any whole number increment up to 189 respectively that includes at least at least 1,2, 3,4,5,6,7, 8, 9,10,11, 12,13, 14,15, 16,17,18,19,20,21, 22,23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; (XXI) a polypeptide comprising at least 5, 6,7,8,9, 10,11,12,13,14,15,16,17,18,19,20,21, 22,23,24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3C, 3E-3L in any whole number increment up to 189 respectively that includes at least at least 1,2, 3,4, 5, 6,7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XXII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12,13, 14, 15, 16,17, 18,19,20,21,22,23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3C, 3E-3L in any whole number increment up to 189 respectively that includes at least at least 1,2, 3,4,5,6,7, 8, 9,10, 11,12,13, 14,15, 16, 17, 18,19, 20,21,22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; (XXIII) a polypeptide comprising at least 5, 6,7,8,9, 10,11,12,13,14,15,16,17,18,19, 20,21,22,23, 24, 26, 27, 28, 29, 30, 31,32, 33, 34, amino acids of a protein of Figure 3C, 3E-3L in any whole number increment up to 189 respectively that includes at least at least 1,2, 3,4, 5,6,7, 8, 9, 10, 11, 12,13, 14,15, 16,17,18,19, 20,21,22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid positon(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XXIV) a polypeptide comprising at least 5, 6, 7, 8, 9, 10,11,12,13,14,15,16,17, 18,19,20,21, 22, 23, 24, 26, 27, 28, 29, 30, 31,32, 33, 34, 35 amino acids of a protein of Figure 3D, in any whole number increment up to 114 respectively that includes at least 1, 2,3,4, 5,6,7, 8,9,10, 11,12,13,14, 15, 16,17,18, 19, 20,21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of Figure (XXV) a polypeptide comprising at least 5, 6,7,8,9,10,11,12,13, 14,15,16,17,18,19,20,21, 22,23, 24, 26, 27, 28, 29, 30, 31,32, 33, 34, 35 amino acids of a protein of Figure 3D, in any whole number increment up to 114 respectively that includes at least at least 1,2, 3, 4, 5, 6, 7, 8, 9,10, 11,12, 13,14, 15,16, 17,18,19, 20, 21,22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of Figure 6; WO 2005/014780 PCT/US2004/017231 (XXVI) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12,13,14,15,16,17, 18,19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3D, in any whole number increment up to 114 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11,12, 13,14, 15, 16,17,18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; (XXVII) a polypeptide comprising at least 5, 6, 7, 8, 9,10,11, 12,13,14,15,16,17, 18,19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of Figure 3D, in any whole number increment up to 114 respectively that includes at least atleast 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11,12, 13,14, 15,16, 17,18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of Figure 8; (XXVIII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12,13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, amino acids of a protein of Figure 3D in any whole number increment up to 114 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of Figure 9; (XXIX) a peptide that occurs at least twice in Tables VIII-XXI and XXII to XLIX, collectively; (XXX) a peptide that occurs at least three times in Tables VIII-XXI and XXII to XLIX, collectively; (XXXI) a peptide that occurs at least four times in Tables VIII-XXI and XXII to XLIX, collectively; (XXXII) a peptide that occurs at least five times in Tables VIII-XXI and XXII to XLIX, collectively; (XXXIII) a peptide that occurs at least once in Tables VIII-XXI, and at least once in tables XXII to XLIX; (XXXIV) a peptide that occurs at least once in Tables VIII-XXI, and at least twice in tables XXII to XLIX; (XXXV) a peptide that occurs at least twice in Tables VIII-XXI, and at least once in tables XXII to XLIX; (XXXVI) a peptide that occurs at least twice in Tables VIII-XXI, and at least twice in tables XXII to XLIX; (XXXVII) a peptide which comprises one two, three, four, or five of the following characteristics, or an oligonucleotide encoding such peptide: i) a region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Hydrophilicity profile of Figure ii) a region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or less than 0.4, 0.3, 0.2, 0.1, or having a value equal to 0.0, in the Hydropathicity profile of Figure 6; iii) a region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Percent Accessible Residues profile of Figure 7; iv) a region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Average Flexibility profile of Figure 8; or, v) a region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Beta-turn profile of Figure 9; 32 WO 2005/014780 PCT/US2004/017231 (XXXVIII) a composition comprising a peptide of (I)-(XXXVII) or an antibody or binding region thereof together with a pharmaceutical excipient and/or in a human unit dose form; (XXXIX) a method of using a peptide of (I)-(XXXVII), or an antibody or binding region thereof or a composition of (XXXVIII) in a method to modulate a cell expressing PSCA; (XL) a method of using a peptide of (I)-(XXXVII) or an antibody or binding region thereof or a composition of (XXXVIII) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing PSCA; (XLI) a method of using a peptide of (I)-(XXXVII) or an antibody or binding region thereof or a composition (XXXVIII) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing PSCA, said cell from a cancer of a tissue listed in Table I; (XLII) a method of using a peptide of (I)-(XXXVII) or an antibody or binding region thereof or a composition of (XXXVIII) in a method to diagnose, prophylax, prognose, or treat a a cancer; (XLIII) a method of using a peptide of (I)-(XXXVII) or an antibody or binding region thereof or a composition of (XXXVIII) in a method to diagnose, prophylax, prognose, or treat a a cancer of a tissue listed in Table I; and; (XLIV) a method of using a a peptide of (I)-(XXXVII) or an antibody or binding region thereof or a composition (XXXVIII) in a method to identify or characterize a modulator of a cell expressing PSCA.
[0120] As used herein, a range is understood to specifically disclose all whole unit positions thereof.
[0121] Typical embodiments of the invention disclosed herein include PSCA polynucleotides that encode specific portions of PSCA mRNA sequences (and those which are complementary to such sequences) such as those that encode the proteins and/or fragments thereof, for example: 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 80, 85, 90, 95, 100, 105, 110, 115, 120, and 123 or more contiguous amino acids of PSCA variant 1; the maximal lengths relevant for other variants are: variant 3, 94 amino acids; variant 4, 189 amino acids, variant 6, 114 amino acids, variant 19, 189 amino acids, variant 20, 189 amino acids, variant 21,189 amino acids, variant 22, 189 amino acids, variant 24, 189 amino acids, variant 25, 189 amino acids, variant 26, 189 amino acids, and variant 27, 189 amino acids..
[0122] In general, naturally occurring allelic variants of human PSCA share a high degree of structural identity and homology 90% or more homology). Typically, allelic variants of a PSCA protein contain conservative amino acid substitutions within the PSCA sequences described herein or contain a substitution of an amino acid from a corresponding position in a homologue of PSCA. One class of PSCA allelic variants are proteins that share a high degree of homology with at least a small region of a particular PSCA amino acid sequence, but further contain a radical departure from the sequence, such as a non-conservative substitution, truncation, insertion or frame shift. In comparisons of protein sequences, the terms, similarity, identity, and homology each have a distinct meaning as appreciated in the field of genetics. Moreover, orthology and paralogy can be important concepts describing the relationship of members of a given protein family in one organism to the members of the same family in other organisms.
[0123] Amino acid abbreviations are provided in Table II. Conservative amino acid substitutions can frequently be made in a protein without altering either the conformation or the function of the protein. Proteins of the invention can comprise 1, 2, 3, 4, 5, 6, 7, 8, 9,10, 11, 12, 13,14,15 conservative substitutions. Such changes include substituting any of isoleucine valine and leucine for any other of these hydrophobic amino acids; aspartic acid for glutamic acid and vice versa; glutamine for asparagine and vice versa; and serine for threonine and vice versa. Other substitutions can also be considered conservative, depending on the environment of the particular amino acid and its role in the three-dimensional structure of the protein. For example, glycine and alanine can frequently be interchangeable, 33 WO 2005/014780 PCT/US2004/017231 as can alanine and valine Methionine which is relatively hydrophobic, can frequently be interchanged with leucine and isoleucine, and sometimes with valine. Lysine and arginine are frequently interchangeable in locations in which the significant feature of the amino acid residue is its charge and the differing pK's of these two amino acid residues are not significant. Still other changes can be considered "conservative" in particular environments (see, e.g.
Table III herein; pages 13-15 "Biochemistry" 2nd ED. Lubert Stryer ed (Stanford University); Henikoff et al, PNAS 1992 Vol 89 10915-10919; Lei etal., J Biol Chem 1995 May 19; 270(20):11882-6).
[0124] Embodiments of the invention disclosed herein include a wide variety of art-accepted variants or analogs of PSCA proteins such as polypeptides having amino acid insertions, deletions and substitutions. PSCA variants can be made using methods known in the art such as site-directed mutagenesis, alanine scanning, and PCR mutagenesis. Sitedirected mutagenesis (Carter et al, Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl. Acids Res., 10:6487 (1987)), cassette mutagenesis (Wells et Gene, 34:315 (1985)), restriction selection mutagenesis (Wells et al., Philos. Trans. R.
Soc. London SerA, 317:415 (1986)) or other known techniques can be performed on the cloned DNA to produce the PSCA variant DNA.
[0125] Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence that is involved in a specific biological activity such as a protein-protein interaction. Among the preferred scanning amino acids are relatively small, neutral amino adds. Such amino acids include alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions (Creighton, The Proteins, Freeman Co., Chothia, J. Mol. Biol., 150:1 (1976)). If alanine substitution does not yield adequate amounts of variant, an isosteric amino acid can be used.
[0126] As defined herein, PSCA variants, analogs or homologs, have the distinguishing attribute of having at least one epitope that is "cross reactive" with a PSCA protein having an amino acid sequence of Figure 3. As used in this sentence, "cross reactive" means that an antibody or T cell that specifically binds to a PSCA variant also specifically binds to a PSCA protein having an amino acid sequence set forth in Figure 3. A polypeptide ceases to be a variant of a protein shown in Figure 3, when it no longer contains any epitope capable of being recognized by an antibody or T cell that specifically binds to the starting PSCA protein. Those skilled in the art understand that antibodies that recognize proteins bind to epitopes of varying size, and a grouping of the order of about four or five amino acids, contiguous or not, is regarded as a typical number of amino acids in a minimal epitope. See, Nair et al., J. Immunol 2000 165(12): 6949-6955; Hebbes et a, Mol Immunol (1989) 26(9):865-73; Schwartz et al., J Immunol (1985) 135(4):2598-608.
[0127] Other classes of PSCA-related protein variants share 70%, 75%, 80%, 85% or 90% or more similarity with an amino acid sequence of Figure 3, or a fragment thereof. Another specific class of PSCA protein variants or analogs comprises one or more of the PSCA biological motifs described herein or presently known in the art. Thus, encompassed by the present invention are analogs of PSCA fragments (nucleic or amino acid) that have altered functional (e.g.
immunogenic) properties relative to the starting fragment. It is to be appreciated that motifs now or which become part of the art are to be applied to the nucleic or amino acid sequences of Figure 2 or Figure 3.
[0128] As discussed herein, embodiments of the claimed invention include polypeptides containing less than the full amino acid sequence of a PSCA protein shown in Figure 2 or Figure 3. For example, representative embodiments of the invention comprise peptideslproteins having any4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more contiguous amino acids of a PSCA protein shown in Figure 2 or Figure 3.
WO 2005/014780 PCT/US2004/017231 [0129] Moreover, representative embodiments of the invention disclosed herein include polypeptides consisting of about amino acid 1 to about amino acid 10 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 10 to about amino acid 20 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 20 to about amino acid 30 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 30 to about amino acid 40 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 40 to about amino acid 50 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 50 to about amino acid 60 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 60 to about amino acid 70 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 70 to about amino acid 80 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 80 to about amino acid 90 of a PSCA protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 90 to about amino acid 100 of a PSCA protein shown in Figure 2 or Figure 3, etc. throughout the entirety of a PSCA amino acid sequence. Moreover, polypeptides consisting of about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 130, or 140 or 150 etc.) of a PSCA protein shown in Figure 2 or Figure 3 are embodiments of the invention. It is to be appreciated that the starting and stopping positions in this paragraph refer to the specified position as well as that position plus or minus 5 residues.
[0130] PSCA-related proteins are generated using standard peptide synthesis technology or using chemical cleavage methods well known in the art. Alternatively, recombinant methods can be used to generate nucleic acid molecules that encode a PSCA-related protein. In one embodiment, nucleic acid molecules provide a means to generate defined fragments of a PSCA protein (or variants, homologs or analogs thereof).
III.A.) Motif-bearing Protein Embodiments [0131] Additional illustrative embodiments of the invention disclosed herein include PSCA polypeptides comprising the amino acid residues of one or more of the biological motifs contained within a PSCA polypeptide sequence set forth in Figure 2 or Figure 3. Various motifs are known in the art, and a protein can be evaluated for the presence of such motifs by a number of publicly available Internet sites (see, URL addresses: pfam.wustl.edu/; searchlauncher.bcm.tmc.edu/seqsearch/struc-predict.html; psort.ims.u-tokyo.ac.jp/; cbs.dtu.dk/; ebi.ac.uklinterprolscan.html; expasy.ch/tools/scnpsitl.html; Epimatrix TM and EpimerM, Brown University, brown.edu/ResearchlTB-HIVLablepimatrix/epimatrix.html; and BIMAS, bimas.dcrt.nih.gov/.).
[0132] Motif bearing subsequences of all PSCA variant proteins are set forth and identified in Tables VIII-XXI and
XXII-XLIX.
[0133] Table V sets forth several frequently occurring motifs based on pfam searches (see URL address pfam.wustl.edu/). The columns of Table V list motif name abbreviation, percent identity found amongst the different member of the motif family, motif name or description and most common function; location information is included if the motif is relevant for location.
[0134] Polypeptides comprising one or more of the PSCA motifs discussed above are useful in elucidating the specific characteristics of a malignant phenotype in view of the observation that the PSCA motifs discussed above are associated with growth dysregulation and because PSCA is overexpressed in certain cancers (See, Table Casein kinase II, cAMP and camp-dependent protein kinase, and Protein Kinase C, for example, are enzymes known to be associated with the development of the malignant phenotype (see e.g. Chen et Lab Invest., 78(2): 165-174 (1998); Gaiddon et al., Endocrinology 136(10): 4331-4338 (1995); Hall et al, Nucleic Acids Research 24(6): 1119-1126 (1996); WO 2005/014780 PCT/US2004/017231 Peterziel et at., Oncogene 18(46): 6322-6329 (1999) and O'Brian, Oncol. Rep. 305-309 (1998)). Moreover, both glycosylation and myristoylation are protein modifications also associated with cancer and cancer progression (see e.g.
Dennis etal., Biochem. Biophys. Acta 1473(1):21-34 (1999); Raju etal., Exp. Cell Res. 235(1): 145-154 (1997)). Amidation is another protein modification also associated with cancer and cancer progression (see e.g. Treston et al, J. Natl. Cancer Inst. Monogr. 169-175 (1992)).
[0135] In another embodiment, proteins of the invention comprise one or more of the immunoreactive epitopes identified in accordance with art-accepted methods, such as the peptides set forth in Tables VIII-XXI and XXII-XLIX. CTL epitopes can be determined using specific algorithms to identify peptides within a PSCA protein that are capable of optimally binding to specified HLA alleles Table IV; EpimatrixTM and EpimerTM, Brown University, URL brown.edu/Research/TB- HIVLab/epimatrix/epimatrix.html; and BIMAS, URL bimas.dcrt.nih.gov/.) Moreover, processes for identifying peptides that have sufficient binding affinity for HLA molecules and which are correlated with being immunogenic epitopes, are well known in the art, and are carried out without undue experimentation. In addition, processes for identifying peptides that are immunogenic epitopes, are well known in the art, and are carried out without undue experimentation either in vitro or in vivo.
[0136] Also known in the art are principles for creating analogs of such epitopes in order to modulate immunogenicity. For example, one begins with an epitope that bears a CTL or HTL motif (see, the HLA Class I and HLA Class II motifs/supermotifs of Table IV). The epitope is analoged by substituting out an amino acid at one of the specified positions, and replacing it with another amino acid specified for that position. For example, on the basis of residues defined in Table IV, one can substitute out a deleterious residue In favor of any other residue, such as a preferred residue; substitute a less-preferred residue with a preferred residue; or substitute an originally-occurring preferred residue with another preferred residue. Substitutions can occur at primary anchor positions or at other positions in a peptide; see, Table IV.
[0137] A variety of references reflect the art regarding the identification and generation of epitopes in a protein of interest as well as analogs thereof. See, for example, WO 97/33602 to Chesnut et al.; Sette, Immunogenetics 1999 50(3- 201-212; Sette et al., J. Immunol. 2001 166(2): 1389-1397; Sidney et Hum. Immunol. 1997 58(1): 12-20; Kondo et al, Immunogenetics 1997 45(4): 249-258; Sidney et al., J. Immunol. 1996 157(8): 3480-90; and Falk etal., Nature 351: 290-6 (1991); Hunt et al., Science 255:1261-3 (1992); Parker et al., J. Immunol. 149:3580-7 (1992); Parker et J.
Immunol. 152:163-75 (1994)); Kast et al., 1994 152(8): 3904-12; Borras-Cuesta etal, Hum. Immunol. 2000 61(3): 266-278; Alexander et aL, J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et at, PMID: 7895164, UI: 95202582; O'Sullivan et J. Immunol. 1991 147(8): 2663-2669; Alexander et al., Immunity 1994 751-761 and Alexander et at, Immunol.
Res. 1998 18(2): 79-92.
[0138] Related embodiments of the invention include polypeptides comprising combinations of the different motifs set forth in Table VI, and/or, one or more of the predicted CTL epitopes of Tables VIII-XXI and XXII-XLIX, and/or, one or more of the predicted HTL epitopes of Tables XLVI-XLIX, and/or, one or more of the T cell binding motifs known in the art.
Preferred embodiments contain no insertions, deletions or substitutions either within the motifs or within the intervening sequences of the polypeptides. In addition, embodiments which include a number of either N-terminal and/or C-terminal amino acid residues on either side of these motifs may be desirable (to, for example, include a greater portion of the polypeptide architecture in which the motif is located). Typically, the number of N-terminal and/or C-terminal amino acid residues on either side of a motif is between about 1 to about 100 amino acid residues, preferably 5 to about 50 amino acid residues.
WO 2005/014780 PCT/US2004/017231 [0139] PSCA-related proteins are embodied in many forms, preferably in isolated form. A purified PSCA protein molecule will be substantially free of other proteins or molecules that impair the binding of PSCA to antibody, T cell or other ligand. The nature and degree of isolation and purification will depend on the intended use. Embodiments of a PSCA-related proteins include purified PSCA-related proteins and functional, soluble PSCA-related proteins, In one embodiment, a functional, soluble PSCA protein or fragment thereof retains the ability to be bound by antibody, T cell or other ligand.
[0140] The invention also provides PSCA proteins comprising biologically active fragments of a PSCA amino acid sequence shown in Figure 2 or Figure 3. Such proteins exhibit properties of the starting PSCA protein, such as the ability to elicit the generation of antibodies that specifically bind an epitope associated with the starting PSCA protein; to be bound by such antibodies; to elicit the activation of HTL or CTL; and/or, to be recognized by HTL or CTL that also specifically bind to the starting protein.
[0141] PSCA-related polypeptides that contain particularly interesting structures can be predicted and/or identified using various analytical techniques well known in the art, including, for example, the methods of Chou-Fasman, Garnier- Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis, or based on immunogenicity. Fragments that contain such structures are particularly useful in generating subunit-specific anti-PSCA antibodies or T cells or in identifying cellular factors that bind to PSCA. For example, hydrophilicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Hopp, T.P. and Woods, 1981, Proc. Natl. Acad. Sci. U.S.A.
78:3824-3828. Hydropathicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Kyte, J. and Doolittle, 1982, J. Mol. Biol. 157:105-132. Percent Accessible Residues profiles can be generated, and immunogenic peptide fragments identified, using the method of Janin 1979, Nature 277:491-492. Average Flexibility profiles can be generated, and immunogenic peptide fragments identified, using the method of Bhaskaran R., Ponnuswamy 1988, Int. J. Pept. Protein Res. 32:242-255. Beta-turn profiles can be generated, and immunogenic peptide fragments identified, using the method of Deleage, Roux 1987, Protein Engineering 1:289-294.
[0142] CTL epitopes can be determined using specific algorithms to identify peptides within a PSCA protein that are capable of optimally binding to specified HLA alleles by using the SYFPEITHI site at World Wide Web URL syfpeithi.bmi-heidelberg.com/; the listings in Table Epimatrix T M and Epimer T M Brown University, URL (brown.edu/Research/TB-HIVLab/epimatrix/epimatrix.html); and BIMAS, URL bimas.dcrt.nih.gov/). Illustrating this, peptide epitopes from PSCA that are presented in the context of human MHC Class I molecules, HLA-A1, A2, A3, Ali, A24, B7 and B35 were predicted (see, Tables VIII-XXI, XXII-XLIX). Specifically, the complete amino acid sequence of the PSCA protein and relevant portions of other variants, for HLA Class I predictions 9 flanking residues on either side of a point mutation or exon juction, and for HLA Class II predictions 14 flanking residues on either side of a point mutation or exon junction corresponding to that variant, were entered into the HLA Peptide Motif Search algorithm found in the Bioinformatics and Molecular Analysis Section (BIMAS) web site listed above; in addition to the site SYFPEITHI, at URL syfpeithi.bmi-heidelberg.coml.
[0143] The HLA peptide motif search algorithm was developed by Dr. Ken Parker based on binding of specific peptide sequences in the groove of HLA Class I molecules, in particular HLA-A2 (see, Falk et Nature 351: 290-6 (1991); Hunt et Science 255:1261-3 (1992); Parker et J. Immunol. 149:3580-7 (1992); Parker et al, J. Immunol.
152:163-75 (1994)). This algorithm allows location and ranking of 8-mer, 9-mer, and 10-mer peptides from a complete protein sequence for predicted binding to HLA-A2 as well as numerous other HLA Class I molecules. Many HLA class I binding peptides are 10 or 11-mers. For example, for Class I HLA-A2, the epitopes preferably contain a leucine (L) or methionine at position 2 and a valine or leucine at the C-terminus (see, Parker et al., J. Immunol.
37 WO 2005/014780 PCT/US2004/017231 149:3580-7 (1992)). Selected results of PSCA predicted binding peptides are shown in Tables VIII-XXI and XXII-XLIX herein. In Tables VIII-XXI and XXII-XLVII, selected candidates, 9-mers and 10-mers, for each family member are shown along with their location, the amino acid sequence of each specific peptide, and an estimated binding score. In Tables XLVI-XLIX, selected candidates, 15-mers, for each family member are shown along with their location, the amino acid sequence of each specific peptide, and an estimated binding score. The binding score corresponds to the estimated half time of dissociation of complexes containing the peptide at 37oC at pH 6.5. Peptides with the highest binding score are predicted to be the most tightly bound to HLA Class I on the cell surface for the greatest period of time and thus represent the best immunogenic targets for T-cell recognition.
[0144] Actual binding of peptides to an HLA allele can be evaluated by stabilization of HLA expression on the antigen-processing defective cell line T2 (see, Xue etal, Prostate 30:73-8 (1997) and Peshwa etal, Prostate 36:129- 38 (1998)). Immunogenicity of specific peptides can be evaluated in vitro by stimulation of CD8+ cytotoxic T lymphocytes (CTL) in the presence of antigen presenting cells such as dendritic cells.
[0145] It is to be appreciated that every epitope predicted by the BIMAS site, EpimerTM and EpimatrixT sites, or specified by the HLA class I or class II motifs available in the art or which become part of the art such as set forth in Table IV (or determined using World Wide Web site URL syfpeithi.bmi-heidelberg.com/, or BIMAS, bimas.dcrt.nih.gov/) are to be "applied" to a PSCA protein in accordance with the invention. As used in this context "applied" means that a PSCA protein is evaluated, visually or by computer-based patterns finding methods, as appreciated by those of skill in the relevant art. Every subsequence of a PSCA protein of 8, 9, 10, or 11 amino acid residues that bears an HLA Class I motif, or a subsequence of 9 or more amino acid residues that bear an HLA Class II motif are within the scope of the invention.
IIl.BI) Expression of PSCA-related Proteins [0146] In an embodiment described in the examples that follow, PSCA can be conveniently expressed in cells (such as 293T cells) transfected with a commercially available expression vector such as a CMV-driven expression vector encoding PSCA with a C-terminal 6XHis and MYC tag (pcDNA3.1/mycHIS, Invitrogen or Tag5, GenHunter Corporation, Nashville TN). The Tag5 vector provides an IgGK secretion signal that can be used to facilitate the production of a secreted PSCA protein in transfected cells. The secreted HIS-tagged PSCA in the culture media can be purified, e.g., using a nickel column using standard techniques.
III.C.) Modifications of PSCA-related Proteins [0147] Modifications of PSCA-related proteins such as covalent modifications are included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of a PSCA polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of a PSCA protein. Another type of covalent modification of a PSCA polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of a protein of the invention. Another type of covalent modification of PSCA comprises linking a PSCA polypeptide to one of a variety of nonproteinaceous polymers, polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.
[0148] The PSCA-related proteins of the present invention can also be modified to form a chimeric molecule comprising PSCA fused to another, heterologous polypeptide or amino acid sequence. Such a chimeric molecule can be WO 2005/014780 PCT/US2004/017231 synthesized chemically or recombinantly. A chimeric molecule can have a protein of the invention fused to another tumorassociated antigen or fragment thereof. Alternatively, a protein in accordance with the invention can comprise a fusion of fragments of a PSCA sequence (amino or nucleic acid) such that a molecule is created that is not, through its length, directly homologous to the amino or nucleic acid sequences shown in Figure 2 or Figure 3. Such a chimeric molecule can comprise multiples of the same subsequence of PSCA. A chimeric molecule can comprise a fusion of a PSCA-related protein with a polyhistidine epitope tag, which provides an epitope to which immobilized nickel can selectively bind, with cytokines or with growth factors. The epitope tag is generally placed at the amino- or carboxyl- terminus of a PSCA protein.
In an alternative embodiment, the chimeric molecule can comprise a fusion of a PSCA-related protein with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimeric molecule (also referred to as an "immunoadhesin"), such a fusion could be to the Fc region of an IgG molecule. The Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of a PSCA polypeptide in place of at least one variable region within an Ig molecule. In a preferred embodiment, the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CHI, CH2 and CH3 regions of an IgGI molecule. For the production of immunoglobulin fusions see, e.g., U.S. Patent No. 5,428,130 issued June 27,1995.
III.D.) Uses of PSCA-related Proteins [0149] The proteins of the invention have a number of different specific uses. As PSCA is highly expressed in prostate and other cancers, PSCA-related proteins are used in methods that assess the status of PSCA gene products in normal versus cancerous tissues, thereby elucidating the malignant phenotype. Typically, polypeptides from specific regions of a PSCA protein are used to assess the presence of perturbations (such as deletions, insertions, point mutations etc.) in those regions (such as regions containing one or more motifs). Exemplary assays utilize antibodies or T cells targeting PSCA-related proteins comprising the amino acid residues of one or more of the biological motifs contained within a PSCA polypeptide sequence in order to evaluate the characteristics of this region in normal versus cancerous tissues or to elicit an immune response to the epitope. Alternatively, PSCA-related proteins that contain the amino acid residues of one or more of the biological motifs in a PSCA protein are used to screen for factors that interact with that region of PSCA.
[0150] PSCA protein fragments/subsequences are particularly useful in generating and characterizing domainspecific antibodies antibodies recognizing an extracellular or intracellular epitope of a PSCA protein), for identifying agents or cellular factors that bind to PSCA or a particular structural domain thereof, and in various therapeutic and diagnostic contexts, including but not limited to diagnostic assays, cancer vaccines and methods of preparing such vaccines.
[0151] Proteins encoded by the PSCA genes, or by analogs, homologs or fragments thereof, have a variety of uses, including but not limited to generating antibodies and in methods for identifying ligands and other agents and cellular constituents that bind to a PSCA gene product. Antibodies raised against a PSCA protein or fragment thereof are useful in diagnostic and prognostic assays, and imaging methodologies in the management of human cancers characterized by expression of PSCA protein, such as those listed in Table I. Such antibodies can be expressed intracellularly and used in methods of treating patients with such cancers. PSCA-related nucleic acids or proteins are also used in generating HTL or CTL responses.
[0152] Various immunological assays useful for the detection of PSCA proteins are used, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), immunocytochemical methods, and the like. Antibodies can be labeled and used as immunological 39 WO 2005/014780 PCT/US2004/017231 imaging reagents capable of detecting PSCA-expressing cells in radioscintigraphic imaging methods). PSCA proteins are also particularly useful in generating cancer vaccines, as further described herein.
IV.) PSCA Antibodies [0153] Another aspect of the invention provides antibodies that bind to PSCA-related proteins. Preferred antibodies specifically bind to a PSCA-related protein and do not bind (or bind weakly) to peptides or proteins that are not PSCArelated proteins under physiological conditions. In this context, examples of physiological conditions include: 1) phosphate buffered saline; 2) Tris-buffered saline containing 25mM Tris and 150 mM NaCI; or normal saline NaCI); 4) animal serum such as human serum; or, 5) a combination of any of 1) through these reactions preferably taking place at pH alternatively in a range of pH 7.0 to 8.0, or alternatively in a range of pH 6.5 to 8.5; also, these reactions taking place at a temperature between 4 0 C to 37"C. For example, antibodies that bind PSCA can bind PSCA-related proteins such as the homologs or analogs thereof, [0154] PSCA antibodies of the invention are particularly useful in cancer (see, Table I) diagnostic and prognostic assays, and imaging methodologies. Similarly, such antibodies are useful in the treatment, diagnosis, and/or prognosis of other cancers, to the extent PSCA is also expressed or overexpressed in these other cancers. Moreover, intracellularly expressed antibodies single chain antibodies) are therapeutically useful in treating cancers in which the expression of PSCA is involved, such as advanced or metastatic prostate cancers.
[0155] The invention also provides various immunological assays useful for the detection and quantification of PSCA and mutant PSCA-related proteins. Such assays can comprise one or more PSCA antibodies capable of recognizing and binding a PSCA-related protein, as appropriate. These assays are performed within various immunological assay formats well known in the art, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), and the like.
[0156] Immunological non-antibody assays of the invention also comprise T cell immunogenicity assays (inhibitory or stimulatory) as well as major histocompatibility complex (MHC) binding assays.
[0157] In addition, immunological imaging methods capable of detecting prostate cancer and other cancers expressing PSCA are also provided by the invention, including but not limited to radioscintigraphic imaging methods using labeled PSCA antibodies. Such assays are clinically useful in the detection, monitoring, and prognosis of PSCA expressing cancers such as prostate cancer.
[0158] PSCA antibodies are also used in methods for purifying a PSCA-related protein and for isolating PSCA homologues and related molecules. For example, a method of purifying a PSCA-related protein comprises incubating a PSCA antibody, which has been coupled to a solid matrix, with a lysate or other solution containing a PSCA-related protein under conditions that permit the PSCA antibody to bind to the PSCA-related protein; washing the solid matrix to eliminate impurities; and eluting the PSCA-related protein from the coupled antibody. Other uses of PSCA antibodies in accordance with the invention include generating anti-idiotypic antibodies that mimic a PSCA protein.
[0159] Various methods for the preparation of antibodies are well known in the art. For example, antibodies can be prepared by immunizing a suitable mammalian host using a PSCA-related protein, peptide, or fragment, in isolated or immunoconjugated form (Antibodies: A Laboratory Manual, CSH Press, Eds., Harlow, and Lane (1988); Harlow, Antibodies, Cold Spring Harbor Press, NY (1989)). In addition, fusion proteins of PSCA can also be used, such as a PSCA GST-fusion protein. In a particular embodiment, a GST fusion protein comprising all or most of the amino acid sequence of Figure 2 or WO 2005/014780 PCT/US2004/017231 Figure 3 is produced, then used as an immunogen to generate appropriate antibodies. In another embodiment, a PSCArelated protein is synthesized and used as an immunogen.
[0160] In addition, naked DNA immunization techniques known in the art are used (with or without purified PSCArelated protein or PSCA expressing cells) to generate an immune response to the encoded immunogen (for review, see Donnelly et al., 1997, Ann. Rev. Immunol. 15: 617-648).
[0161] The amino acid sequence of a PSCA protein as shown in Figure 2 or Figure 3 can be analyzed to select specific regions of the PSCA protein for generating antibodies. For example, hydrophobicity and hydrophilicity analyses of a PSCA amino acid sequence are used to identify hydrophilic regions in the PSCA structure. Regions of a PSCA protein that show immunogenic structure, as well as other regions and domains, can readily be identified using various other methods known in the art, such as Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson- Wolf analysis. Hydrophilicity profiles can be generated using the method of Hopp, T.P. and Woods, 1981, Proc. Natl.
Acad. Sci. U.S.A. 78:3824-3828. Hydropathicity profiles can be generated using the method of Kyte, J. and Doolittle, R.F., 1982, J. Mol. Biol. 157:105-132. Percent Accessible Residues profiles can be generated using the method of Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be generated using the method of Bhaskaran Ponnuswamy 1988, Int. J. Pept. Protein Res. 32:242-255. Beta-turn profiles can be generated using the method of Deleage, G., Roux 1987, Protein Engineering 1:289-294. Thus, each region identified by any of these programs or methods is within the scope of the present invention. Methods for the generation of PSCA antibodies are further illustrated by way of the examples provided herein. Methods for preparing a protein or polypeptide for use as an immunogen are well known in the art. Also well known in the art are methods for preparing immunogenic conjugates of a protein with a carrier, such as BSA, KLH or other carrier protein. In some circumstances, direct conjugation using, for example, carbodiimide reagents are used; in other instances linking reagents such as those supplied by Pierce Chemical Co., Rockford, IL, are effective.
Administration of a PSCA immunogen is often conducted by injection over a suitable time period and with use of a suitable adjuvant, as is understood in the art. During the immunization schedule, titers of antibodies can be taken to determine adequacy of antibody formation.
[0162] PSCA monoclonal antibodies can be produced by various means well known in the art. For example, immortalized cell lines that secrete a desired monoclonal antibody are prepared using the standard hybridoma technology of Kohler and Milstein or modifications that immortalize antibody-producing B cells, as is generally known, Immortalized cell lines that secrete the desired antibodies are screened by immunoassay in which the antigen is a PSCA-related protein.
When the appropriate immortalized cell culture is identified, the cells can be expanded and antibodies produced either from in vitro cultures or from ascites fluid.
[0163] The antibodies or fragments of the invention can also be produced, by recombinant means. Regions that bind specifically to the desired regions of a PSCA protein can also be produced in the context of chimeric or complementarity-determining region (CDR) grafted antibodies of multiple species origin. Humanized or human PSCA antibodies can also be produced, and are preferred for use in therapeutic contexts. Methods for humanizing murine and other non-human antibodies, by substituting one or more of the non-human antibody CDRs for corresponding human antibody sequences, are well known (see for example, Jones etal., 1986, Nature 321: 522-525; Riechmann et 1988, Nature 332: 323-327; Verhoeyen et al., 1988, Science 239: 1534-1536). See also, Carter et al, 1993, Proc. Natl. Acad.
Sci. USA 89: 4285 and Sims et al, 1993, J. Immunol. 151: 2296.
[0164] Methods for producing fully human monoclonal antibodies include phage display and transgenic methods (for review, see Vaughan et al., 1998, Nature Biotechnology 16: 535-539). Fully human PSCA monoclonal antibodies can be 41 WO 2005/014780 PCT/US2004/017231 generated using cloning technologies employing large human Ig gene combinatorial libraries phage display) (Griffiths and Hoogenboom, Building an in vitro immune system: human antibodies from phage display libraries. In: Protein Engineering of Antibody Molecules for Prophylactic and Therapeutic Applications in Man, Clark, M. Nottingham Academic, pp 45-64 (1993); Burton and Barbas, Human Antibodies from combinatorial libraries. Id., pp 65-82). Fully human PSCA monoclonal antibodies can also be produced using transgenic mice engineered to contain human immunoglobulin gene loci as described in PCT Patent Application W09824893, Kucherlapati and Jakobovits et al., published December 3, 1997 (see also, Jakobovits, 1998, Exp. Opin. Invest. Drugs 607-614; U.S. patents 6,162,963 issued 19 December 2000; 6,150,584 issued 12 November 2000; and, 6,114598 issued 5 September 2000). This method avoids the in vitro manipulation required with phage display technology and efficiently produces high affinity authentic human antibodies.
[0165] Reactivity of PSCA antibodies with a PSCA-related protein can be established by a number of well known means, including Western blot, immunoprecipitation, ELISA, and FACS analyses using, as appropriate, PSCA-related proteins, PSCA-expressing cells or extracts thereof. A PSCA antibody or fragment thereof can be labeled with a detectable marker or conjugated to a second molecule. Suitable detectable markers include, but are not limited to, a radioisotope, a fluorescent compound, a bioluminescent compound, chemiluminescent compound, a metal chelator or an enzyme.
Further, bi-specific antibodies specific for two or more PSCA epitopes are generated using methods generally known in the art. Homodimeric antibodies can also be generated by cross-linking techniques known in the art Wolff et al., Cancer Res. 53: 2560-2565).
PSCA Cellular Immune Responses [0166] The mechanism by which T cells recognize antigens has been delineated. Efficacious peptide epitope vaccine compositions of the invention induce a therapeutic or prophylactic immune responses in very broad segments of the world-wide population. For an understanding of the value and efficacy of compositions of the invention that induce cellular immune responses, a brief review of immunology-related technology is provided.
[0167] A complex of an HLA molecule and a peptidic antigen acts as the ligand recognized by HLA-restricted T cells (Buus, S. etal., Cell47:1071, 1986; Babbitt, B. P. etal., Nature 317:359, 1985; Townsend, A. and Bodmer, Annu. Rev.
Immunol 7:601, 1989; Germain, R. Annu. Rev. Immunol. 11:403, 1993). Through the study of single amino acid substituted antigen analogs and the sequencing of endogenously bound, naturally processed peptides, critical residues that correspond to motifs required for specific binding to HLA antigen molecules have been identified and are set forth in Table IV (see also, Southwood, et J. Immunol. 160:3363, 1998; Rammensee, et Immunogenetics 41:178, 1995; Rammensee et al., SYFPEITHI, access via World Wide Web at URL (134.2.96.221/scripts.hlaserver.dll/home.htm); Sette, A. and Sidney, J. Curr. Opin. Immunol. 10:478, 1998; Engelhard, V. Curr. Opin. Immunol. 6:13,1994; Sette, A. and Grey, H. Curr. Opin. Immunol. 4:79, 1992; Sinigaglia, F. and Hammer, J. Curr. Biol. 6:52, 1994; Ruppert etal., Cell 74:929-937, 1993; Kondo et at, J. Immunol. 155:4307-4312,1995; Sidney et J. Immunol. 157:3480-3490, 1996; Sidney et al., Human Immunol. 45:79-93, 1996; Sette, A. and Sidney, J. Immunogenetics 1999 Nov; 50(3-4):201-12, Review).
[0168] Furthermore, x-ray crystallographic analyses of HLA-peptide complexes have revealed pockets within the peptide binding cleft/groove of HLA molecules which accommodate, in an allele-specific mode, residues borne by peptide ligands; these residues in turn determine the HLA binding capacity of the peptides in which they are present. (See, e.g., Madden, D.R. Annu. Rev. Immunol. 13:587, 1995; Smith, et Immunity4:203, 1996; Fremont et Immunity 8:305, 1998; Stern etal., Structure 2:245, 1994; Jones, E.Y. Curr. Opin. Immunol. 9:75, 1997; Brown, J. H. et al., Nature 364:33, 42 WO2005/014780 PCT/US2004/017231 1993; Guo, H. C. et Proc. Natl. Acad. Sci. USA 90:8053, 1993; Guo, H. C. et al., Nature 360:364, 1992; Silver, M. L. et al, Nature 360:367,1992; Matsumura, M. et al., Science 257:927, 1992; Madden etal., Cell 70:1035, 1992; Fremont, D. H.
etal., Science 257:919, 1992; Saper, M. Bjorkman, P. J. and Wiley, D. J. Mol. Biol. 219:277, 1991.) [0169] Accordingly, the definition of class I and class II allele-specific HLA binding motifs, or class I or class II supermotifs allows identification of regions within a protein that are correlated with binding to particular HLA antigen(s).
[0170] Thus, by a process of HLA motif identification, candidates for epitope-based vaccines have been identified; such candidates can be further evaluated by HLA-peptide binding assays to determine binding affinity and/or the time period of association of the epitope and its corresponding HLA molecule. Additional confirmatory work can be performed to select, amongst these vaccine candidates, epitopes with preferred characteristics in terms of population coverage, and/or immunogenicity.
[0171] Various strategies can be utilized to evaluate cellular immunogenicity, including: 1) Evaluation of primary T cell cultures from normal individuals (see, Wentworth, P. A. etal., Mol. Immunol.
32:603, 1995; Cells, E. et Proc. Natl. Acad. Sci. USA 91:2105, 1994; Tsai, V. et al, J. Immunol. 158:1796, 1997; Kawashima, I. et al., Human Immunol. 59:1, 1998). This procedure involves the stimulation of peripheral blood lymphocytes (PBL) from normal subjects with a test peptide in the presence of antigen presenting cells in vitro over a period of several weeks. T cells specific for the peptide become activated during this time and are detected using, a lymphokine- or 5 1 Cr-release assay involving peptide sensitized target cells.
2) Immunization of HLA transgenic mice (see, Wentworth, P. A. et at, J. Immunol. 26:97, 1996; Wentworth, P. A. et Int. Immunol. 8:651, 1996; Alexander, J. et J. Immunol. 159:4753, 1997). For example, in such methods peptides in incomplete Freund's adjuvant are administered subcutaneously to HLA transgenic mice. Several weeks following immunization, splenocytes are removed and cultured in vitro in the presence of test peptide for approximately one week. Peptide-specific T cells are detected using, a 51 Cr-release assay involving peptide sensitized target cells and target cells expressing endogenously generated antigen.
3) Demonstration of recall T cell responses from immune individuals who have been either effectively vaccinated and/or from chronically ill patients (see, Rehermann, B. et al., J. Exp. Med. 181:1047, 1995; Doolan, D. L. et a., Immunity 7:97, 1997; Bertoni, R. et aL, J. Clin. Invest. 100:503, 1997; Threlkeld, S. C. et J. Immunol. 159:1648, 1997; Diepolder, H. M. etal., J. Virol. 71:6011, 1997). Accordingly, recall responses are detected by culturing PBL from subjects that have been exposed to the antigen due to disease and thus have generated an immune response "naturally", or from patients who were vaccinated against the antigen. PBL from subjects are cultured in vitro for 1-2 weeks in the presence of test peptide plus antigen presenting cells (APC) to allow activation of "memory" T cells, as compared to "naive" T cells. At the end of the culture period, T cell activity is detected using assays including 51 Cr release involving peptide-sensitized targets, T cell proliferation, or lymphokine release.
VI.) PSCA Transgenic Animals [0172] Nucleic acids that encode a PSCA-related protein can also be used to generate either transgenic animals or "knock out" animals that, in turn, are useful in the development and screening of therapeutically useful reagents. In accordance with established techniques, cDNA encoding PSCA can be used to clone genomic DNA that encodes PSCA.
The cloned genomic sequences can then be used to generate transgenic animals containing cells that express DNA that encode PSCA. Methods for generating transgenic animals, particularly animals such as mice or rats, have become WO 2005/014780 PCT/US2004/017231 conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 issued 12 April 1988, and 4,870,009 issued 26 September 1989. Typically, particular cells would be targeted for PSCA transgene incorporation with tissuespecific enhancers.
[0173] Transgenic animals that include a copy of a transgene encoding PSCA can be used to examine the effect of increased expression of DNA that encodes PSCA. Such animals can be used as tester animals for reagents thought to confer protection from, for example, pathological conditions associated with its overexpression. In accordance with this aspect of the invention, an animal is treated with a reagent and a reduced incidence of a pathological condition, compared to untreated animals that bear the transgene, would indicate a potential therapeutic intervention for the pathological condition.
[0174] Alternatively, non-human homologues of PSCA can be used to construct a PSCA "knock out" animal that has a defective or altered gene encoding PSCA as a result of homologous recombination between the endogenous gene encoding PSCA and altered genomic DNA encoding PSCA introduced into an embryonic cell of the animal. For example, cDNA that encodes PSCA can be used to clone genomic DNA encoding PSCA in accordance with established techniques.
A portion of the genomic DNA encoding PSCA can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration. Typically, several kilobases of unaltered flanking DNA (both at the 5' and 3' ends) are included in the vector (see, Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors). The vector is introduced into an embryonic stem cell line by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected (see, Li et al., Cell, 69:915 (1992)). The selected cells are then injected into a blastocyst of an animal a mouse or rat) to form aggregation chimeras (see, Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J.
Robertson, ed. (IRL, Oxford, 1987), pp. 113-152). A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal, and the embryo brought to term to create a "knock out" animal. Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knock out animals can be characterized, for example, for their ability to defend against certain pathological conditions or for their development of pathological conditions due to absence of a PSCA polypeptide.
VII.) Methods for the Detection of PSCA [0175] Another aspect of the present invention relates to methods for detecting PSCA polynucleotides and PSCArelated proteins, as well as methods for identifying a cell that expresses PSCA. The expression profile of PSCA makes it a diagnostic marker for metastasized disease. Accordingly, the status of PSCA gene products provides information useful for predicting a variety of factors including susceptibility to advanced stage disease, rate of progression, and/or tumor aggressiveness. As discussed in detail herein, the status of PSCA gene products in patient samples can be analyzed by a variety protocols that are well known in the art including immunohistochemical analysis, the variety of Northern blotting techniques including in situ hybridization, RT-PCR analysis (for example on laser capture micro-dissected samples), Western blot analysis and tissue array analysis.
[0176] More particularly, the invention provides assays for the detection of PSCA polynucleotides in a biological sample, such as serum, bone, prostate, and other tissues, urine, semen, cell preparations, and the like. Detectable PSCA polynucleotides include, for example, a PSCA gene or fragment thereof, PSCA mRNA, alternative splice variant PSCA mRNAs, and recombinant DNA or RNA molecules that contain a PSCA polynucleotide. A number of methods for 44 WO 2005/014780 PCT/US2004/017231 amplifying and/or detecting the presence of PSCA polynucleotides are well known in the art and can be employed in the practice of this aspect of the invention.
[0177] In one embodiment, a method for detecting a PSCA mRNA in a biological sample comprises producing cDNA from the sample by reverse transcription using at least one primer; amplifying the cDNA so produced using a PSCA polynucleotides as sense and antisense primers to amplify PSCA cDNAs therein; and detecting the presence of the amplified PSCA cDNA. Optionally, the sequence of the amplified PSCA cDNA can be determined.
[0178] In another embodiment, a method of detecting a PSCA gene in a biological sample comprises first isolating genomic DNA from the sample; amplifying the isolated genomic DNA using PSCA polynucleotides as sense and antisense primers; and detecting the presence of the amplified PSCA gene. Any number of appropriate sense and antisense probe combinations can be designed from a PSCA nucleotide sequence (see, Figure 2) and used for this purpose.
[0179] The invention also provides assays for detecting the presence of a PSCA protein in a tissue or other biological sample such as serum, semen, bone, prostate, urine, cell preparations, and the like. Methods for detecting a PSCA-related protein are also well known and include, for example, immunoprecipitation, immunohistochemical analysis, Western blot analysis, molecular binding assays, ELISA, ELIFA and the like. For example, a method of detecting the presence of a PSCA-related protein in a biological sample comprises first contacting the sample with a PSCA antibody, a PSCA-reactive fragment thereof, or a recombinant protein containing an antigen-binding region of a PSCA antibody; and then detecting the binding of PSCA-related protein in the sample.
[0180] Methods for identifying a cell that expresses PSCA are also within the scope of the invention. In one embodiment, an assay for identifying a cell that expresses a PSCA gene comprises detecting the presence of PSCA mRNA in the cell. Methods for the detection of particular mRNAs in cells are well known and include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled PSCA riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for PSCA, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like).
Alternatively, an assay for identifying a cell that expresses a PSCA gene comprises detecting the presence of PSCArelated protein in the cell or secreted by the cell. Various methods for the detection of proteins are well known in the art and are employed for the detection of PSCA-related proteins and cells that express PSCA-related proteins.
[0181] PSCA expression analysis is also useful as a tool for identifying and evaluating agents that modulate PSCA gene expression. For example, PSCA expression is significantly upregulated in prostate cancer, and is expressed in cancers of the tissues listed in Table I. Identification of a molecule or biological agent that inhibits PSCA expression or over-expression in cancer cells is of therapeutic value. For example, such an agent can be identified by using a screen that quantifies PSCA expression by RT-PCR, nucleic acid hybridization or antibody binding.
VIII.) Methods for Monitoring the Status of PSCA-related Genes and Their Products [0182] Oncogenesis is known to be a multistep process where cellular growth becomes progressively dysregulated and cells progress from a normal physiological state to precancerous and then cancerous states (see, Alers et Lab Invest, 77(5): 437-438 (1997) and Isaacs et Cancer Surv. 23: 19-32 (1995)). In this context, examining a biological sample for evidence of dysregulated cell growth (such as aberrant PSCA expression in cancers) allows for early detection of such aberrant physiology, before a pathologic state such as cancer has progressed to a stage that therapeutic options are more limited and or the prognosis is worse. In such examinations, the status of PSCA in a biological sample of interest can be compared, for example, to the status of PSCA in a corresponding normal sample a sample from that individual WO 2005/014780 PCT/US2004/017231 or alternatively another individual that is not affected by a pathology). An alteration in the status of PSCA in the biological sample (as compared to the normal sample) provides evidence of dysregulated cellular growth. In addition to using a biological sample that is not affected by a pathology as a normal sample, one can also use a predetermined normative value such as a predetermined normal level of mRNA expression (see, Grever et al, J. Comp. Neurol. 1996 Dec 9; 376(2): 306-14 and U.S. Patent No. 5,837,501) to compare PSCA status in a sample.
[0183] The term "status" in this context is used according to its art accepted meaning and refers to the condition or state of a gene and its products. Typically, skilled artisans use a number of parameters to evaluate the condition or state of a gene and its products. These include, but are not limited to the location of expressed gene products (including the location of PSCA expressing cells) as well as the level, and biological activity of expressed gene products (such as PSCA mRNA, polynucleotides and polypeptides). Typically, an alteration in the status of PSCA comprises a change in the location of PSCA and/or PSCA expressing cells and/or an increase in PSCA mRNA and/or protein expression.
[0184] PSCA status in a sample can be analyzed by a number of means well known in the art, including without limitation, immunohistochemical analysis, in situ hybridization, RT-PCR analysis on laser capture micro-dissected samples, Western blot analysis, and tissue array analysis. Typical protocols for evaluating the status of a PSCA gene and gene products are found, for example in Ausubel et al. eds., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting) and 18 (PCR Analysis). Thus, the status of PSCA in a biological sample is evaluated by various methods utilized by skilled artisans including, but not limited to genomic Southern analysis (to examine, for example perturbations in a PSCA gene), Northern analysis and/or PCR analysis of PSCA mRNA (to examine, for example alterations in the polynucleotide sequences or expression levels of PSCA mRNAs), and, Western and/or immunohistochemical analysis (to examine, for example alterations in polypeptide sequences, alterations in polypeptide localization within a sample, alterations in expression levels of PSCA proteins and/or associations of PSCA proteins with polypeptide binding partners). Detectable PSCA polynucleotides include, for example, a PSCA gene or fragment thereof, PSCA mRNA, alternative splice variants, PSCA mRNAs, and recombinant DNA or RNA molecules containing a PSCA polynucleotide.
[0185] The expression profile of PSCA makes it a diagnostic marker for local and/or metastasized disease, and provides information on the growth or oncogenic potential of a biological sample. In particular, the status of PSCA provides information useful for predicting susceptibility to particular disease stages, progression, andlor tumor aggressiveness. The invention provides methods and assays for determining PSCA status and diagnosing cancers that express PSCA, such as cancers of the tissues listed in Table I. For example, because PSCA mRNA is so highly expressed in prostate and other cancers relative to normal prostate tissue, assays that evaluate the levels of PSCA mRNA transcripts or proteins in a biological sample can be used to diagnose a disease associated with PSCA dysregulation, and can provide prognostic information useful in defining appropriate therapeutic options.
[0186] The expression status of PSCA provides information including the presence, stage and location of dysplastic, precancerous and cancerous cells, predicting susceptibility to various stages of disease, and/or for gauging tumor aggressiveness. Moreover, the expression profile makes it useful as an imaging reagent for metastasized disease.
Consequently, an aspect of the invention is directed to the various molecular prognostic and diagnostic methods for examining the status of PSCA in biological samples such as those from individuals suffering from, or suspected of suffering from a pathology characterized by dysregulated cellular growth, such as cancer.
[0187] As described above, the status of PSCA in a biological sample can be examined by a number of well-known procedures in the art. For example, the status of PSCA in a biological sample taken from a specific location in the body can 46 WO 2005/014780 PCT/US2004/017231 be examined by evaluating the sample for the presence or absence of PSCA expressing cells those that express PSCA mRNAs or proteins). This examination can provide evidence of dysregulated cellular growth, for example, when PSCA-expressing cells are found in a biological sample that does not normally contain such cells (such as a lymph node), because such alterations in the status of PSCA in a biological sample are often associated with dysregulated cellular growth. Specifically, one indicator of dysregulated cellular growth is the metastases of cancer cells from an organ of origin (such as the prostate) to a different area of the body (such as a lymph node). In this context, evidence of dysregulated cellular growth is important for example because occult lymph node metastases can be detected in a substantial proportion of patients with prostate cancer, and such metastases are associated with known predictors of disease progression (see, Murphy at Prostate 42(4): 315-317 (2000);Su et Semin. Surg. Oncol. 18(1): 17-28 (2000) and Freeman et al., J Urol 1995 Aug 154(2 Pt 1):474-8).
[0188] In one aspect, the invention provides methods for monitoring PSCA gene products by determining the status of PSCA gene products expressed by cells from an individual suspected of having a disease associated with dysregulated cell growth (such as hyperplasia or cancer) and then comparing the status so determined to the status of PSCA gene products in a corresponding normal sample. The presence of aberrant PSCA gene products in the test sample relative to the normal sample provides an indication of the presence of dysregulated cell growth within the cells of the individual.
[0189] In another aspect, the invention provides assays useful in determining the presence of cancer in an individual, comprising detecting a significant increase in PSCA mRNA or protein expression in a test cell or tissue sample relative to expression levels in the corresponding normal cell or tissue. The presence of PSCA mRNA can, for example, be evaluated in tissues including but not limited to those listed in Table I. The presence of significant PSCA expression in any of these tissues is useful to indicate the emergence, presence and/or severity of a cancer, since the corresponding normal tissues do not express PSCA mRNA or express it at lower levels.
[0190] In a related embodiment, PSCA status is determined at the protein level rather than at the nucleic acid level.
For example, such a method comprises determining the level of PSCA protein expressed by cells in a test tissue sample and comparing the level so determined to the level of PSCA expressed in a corresponding normal sample. In one embodiment, the presence of PSCA protein is evaluated, for example, using immunohistochemical methods. PSCA antibodies or binding partners capable of detecting PSCA protein expression are used in a variety of assay formats well known in the art for this purpose.
[0191] In a further embodiment, one can evaluate the status of PSCA nucleotide and amino acid sequences in a biological sample in order to identify perturbations in the structure of these molecules. These perturbations can include insertions, deletions, substitutions and the like. Such evaluations are useful because perturbations in the nucleotide and amino acid sequences are observed in a large number of proteins associated with a growth dysregulated phenotype (see, Marrogi et aL, 1999,,J. Cutan. Pathol. 26(8):369-378). For example, a mutation in the sequence of PSCA may be indicative of the presence or promotion of a tumor. Such assays therefore have diagnostic and predictive value where a mutation in PSCA indicates a potential loss of function or increase in tumor growth.
[0192] A wide variety of assays for observing perturbations in nucleotide and amino acid sequences are well known in the art. For example, the size and structure of nucleic acid or amino acid sequences of PSCA gene products are observed by the Northern, Southern, Western, PCR and DNA sequencing protocols discussed herein. In addition, other methods for observing perturbations in nucleotide and amino acid sequences such as single strand conformation polymorphism analysis are well known in the art (see, U.S. Patent Nos. 5,382,510 issued 7 September 1999, and 5,952,170 issued 17 January 1995).
WO 2005/014780 PCT/US2004/017231 [0193] Additionally, one can examine the methylation status of a PSCA gene in a biological sample. Aberrant demethylation and/or hypermethylation of CpG islands in gene 5' regulatory regions frequently occurs in immortalized and transformed cells, and can result in altered expression of various genes. For example, promoter hypermethylation of the piclass glutathione S-transferase (a protein expressed in normal prostate but not expressed in >90% of prostate carcinomas) appears to permanently silence transcription of this gene and is the most frequently detected genomic alteration in prostate carcinomas (De Marzo et aL, Am. J. Pathol. 155(6): 1985-1992 (1999)). In addition, this alteration is present in at least of cases of high-grade prostatic intraepithelial neoplasia (PIN) (Brooks et Cancer Epidemiol. Biomarkers Prev., 1998, 7:531-536). In another example, expression of the LAGE-I tumor specific gene (which is not expressed in normal prostate but is expressed in 25-50% of prostate cancers) is induced by deoxy-azacytidine in lymphoblastoid cells, suggesting that tumoral expression is due to demethylation (Lethe t al., Int. J. Cancer 76(6): 903-908 (1998)). A variety of assays for examining methylation status of a gene are well known in the art. For example, one can utilize, in Southern hybridization approaches, methylation-sensitive restriction enzymes that cannot cleave sequences that contain methylated CpG sites to assess the methylation status of CpG islands. In addition, MSP (methylation specific PCR) can rapidly profile the methylation status of all the CpG sites present in a CpG island of a given gene. This procedure involves initial modification of DNA by sodium bisulfite (which will convert all unmethylated cytosines to uracil) followed by amplification using primers specific for methylated versus unmethylated DNA. Protocols involving methylation interference can also be found for example in Current Protocols In Molecular Biology, Unit 12, Frederick M. Ausubel et al. eds., 1995.
[0194] Gene amplification is an additional method for assessing the status of PSCA. Gene amplification is measured in a sample directly, for example, by conventional Southern blotting or Northern blotting to quantitate the transcription of mRNA (Thomas, 1980, Proc. Natl. Acad. Sci. USA, 77:5201-5205), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies are employed that recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn are labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
[0195] Biopsied tissue or peripheral blood can be conveniently assayed for the presence of cancer cells using for example, Northern, dot blot or RT-PCR analysis to detect PSCA expression. The presence of RT-PCR amplifiable PSCA mRNA provides an indication of the presence of cancer. RT-PCR assays are well known in the art. RT-PCR detection assays for tumor cells in peripheral blood are currently being evaluated for use in the diagnosis and management of a number of human solid tumors. In the prostate cancer field, these include RT-PCR assays for the detection of cells expressing PSA and PSM (Verkaik etal., 1997, Urol. Res. 25:373-384; Ghossein et 1995, J. Clin. Oncol. 13:1195-2000; Heston et 1995, Clin. Chem. 41:1687-1688).
[0196] A further aspect of the invention is an assessment of the susceptibility that an individual has for developing cancer. In one embodiment, a method for predicting susceptibility to cancer comprises detecting PSCA mRNA or PSCA protein in a tissue sample, its presence indicating susceptibility to cancer, wherein the degree of PSCA mRNA expression correlates to the degree of susceptibility. In a specific embodiment, the presence of PSCA in prostate or other tissue is examined, with the presence of PSCA in the sample providing an indication of prostate cancer susceptibility (or the emergence or existence of a prostate tumor). Similarly, one can evaluate the integrity PSCA nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like. The presence of one or more perturbations in PSCA gene products in the sample is an indication of cancer susceptibility (or the emergence or existence of a tumor).
48 WO 2005/014780 PCT/US2004/017231 [0197] The invention also comprises methods for gauging tumor aggressiveness. In one embodiment, a method for gauging aggressiveness of a tumor comprises determining the level of PSCA mRNA or PSCA protein expressed by tumor cells, comparing the level so determined to the level of PSCA mRNA or PSCA protein expressed in a corresponding normal tissue taken from the same individual or a normal tissue reference sample, wherein the degree of PSCA mRNA or PSCA protein expression in the tumor sample relative to the normal sample indicates the degree of aggressiveness. In a specific embodiment, aggressiveness of a tumor is evaluated by determining the extent to which PSCA is expressed in the tumor cells, with higher expression levels indicating more aggressive tumors. Another embodiment is the evaluation of the integrity of PSCA nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like. The presence of one or more perturbations indicates more aggressive tumors.
[0198] Another embodiment of the invention is directed to methods for observing the progression of a malignancy in an individual over time. In one embodiment, methods for observing the progression of a malignancy in an individual over time comprise determining the level of PSCA mRNA or PSCA protein expressed by cells in a sample of the tumor, comparing the level so determined to the level of PSCA mRNA or PSCA protein expressed in an equivalent tissue sample taken from the same individual at a different time, wherein the degree of PSCA mRNA or PSCA protein expression in the tumor sample over time provides information on the progression of the cancer. In a specific embodiment, the progression of a cancer is evaluated by determining PSCA expression in the tumor cells over time, where increased expression over time indicates a progression of the cancer. Also, one can evaluate the integrity PSCA nucleotide and amino acid sequences in a biological sample in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like, where the presence of one or more perturbations indicates a progression of the cancer.
[0199] The above diagnostic approaches can be combined with any one of a wide variety of prognostic and diagnostic protocols known in the art. For example, another embodiment of the invention is directed to methods for observing a coincidence between the expression of PSCA gene and PSCA gene products (or perturbations in PSCA gene and PSCA gene products) and a factor that is associated with malignancy, as a means for diagnosing and prognosticating the status of a tissue sample. A wide variety of factors associated with malignancy can be utilized, such as the expression of genes associated with malignancy PSA, PSCA and PSM expression for prostate cancer etc.) as well as gross cytological observations (see, Booking etal., 1984, Anal. Quant. Cytol. 6(2):74-88; Epstein, 1995, Hum. Pathol.
26(2):223-9; Thorson etal., 1998, Mod. Pathol. 11(6):543-51; Baisden etal., 1999, Am. J. Surg. Pathol. 23(8):918-24).
Methods for observing a coincidence between the expression of PSCA gene and PSCA gene products (or perturbations in PSCA gene and PSCA gene products) and another factor that is associated with malignancy are useful, for example, because the presence of a set of specific factors that coincide with disease provides information crucial for diagnosing and prognosticating the status of a tissue sample.
[0200] In one embodiment, methods for observing a coincidence between the expression of PSCA gene and PSCA gene products (or perturbations in PSCA gene and PSCA gene products) and another factor associated with malignancy entails detecting the overexpression of PSCA mRNA or protein in a tissue sample, detecting the overexpression of PSA mRNA or protein in a tissue sample (or PSCA or PSM expression), and observing a coincidence of PSCA mRNA or protein and PSA mRNA or protein overexpression (or PSCA or PSM expression). In a specific embodiment, the expression of PSCA and PSA mRNA in prostate tissue is examined, where the coincidence of PSCA and PSA mRNA overexpression in the sample indicates the existence of prostate cancer, prostate cancer susceptibility or the emergence or status of a prostate tumor.
WO 2005/014780 PCT/US2004/017231 [0201] Methods for detecting and quantifying the expression of PSCA mRNA or protein are described herein, and standard nucleic acid and protein detection and quantification technologies are well known in the art. Standard methods for the detection and quantification of PSCA mRNA include in situ hybridization using labeled PSCA riboprobes, Northern blot and related techniques using PSCA polynucleotide probes, RT-PCR analysis using primers specific for PSCA, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like. In a specific embodiment, semi-quantitative RT-PCR is used to detect and quantify PSCA mRNA expression. Any number of primers capable of amplifying PSCA can be used for this purpose, including but not limited to the various primer sets specifically described herein. In a specific embodiment, polyclonal or monoclonal antibodies specifically reactive with the wild-type PSCA protein can be used in an immunohistochemical assay of biopsied tissue.
IX.) Identification of Molecules That Interact With PSCA [0202] The PSCA protein and nucleic acid sequences disclosed herein allow a skilled artisan to identify proteins, small molecules and other agents that interact with PSCA, as well as pathways activated by PSCA via any one of a variety of art accepted protocols. For example, one can utilize one of the so-called interaction trap systems (also referred to as the "two-hybrid assay"). In such systems, molecules interact and reconstitute a transcription factor which directs expression of a reporter gene, whereupon the expression of the reporter gene is assayed. Other systems identify protein-protein interactions in vivo through reconstitution of a eukaryotic transcriptional activator, see, U.S. Patent Nos. 5,955,280 issued 21 September 1999, 5,925,523 issued 20 July 1999, 5,846,722 issued 8 December 1998 and 6,004,746 issued 21 December 1999. Algorithms are also available in the art for genome-based predictions of protein function (see, e.g., Marcotte, etal, Nature 402:4 November 1999, 83-86).
[0203] Alternatively one can screen peptide libraries to identify molecules that interact with PSCA protein sequences.
In such methods, peptides that bind to PSCA are identified by screening libraries that encode a random or controlled collection of amino acids. Peptides encoded by the libraries are expressed as fusion proteins of bacteriophage coat proteins, the bacteriophage particles are then screened against the PSCA protein(s).
[0204] Accordingly, peptides having a wide variety of uses, such as therapeutic, prognostic or diagnostic reagents, are thus identified without any prior information on the structure of the expected ligand or receptor molecule. Typical peptide libraries and screening methods that can be used to identify molecules that interact with PSCA protein sequences are disclosed for example in U.S. Patent Nos. 5,723,286 issued 3 March 1998 and 5,733,731 issued 31 March 1998.
[0205] Alternatively, cell lines that express PSCA are used to identify protein-protein interactions mediated by PSCA.
Such interactions can be examined using immunoprecipitation techniques (see, Hamilton et a. Biochem.
Biophys. Res. Commun. 1999, 261:646-51). PSCA protein can be immunoprecipitated from PSCA-expressing cell lines using anti-PSCA antibodies. Alternatively, antibodies against His-tag can be used in a cell line engineered to express fusions of PSCA and a His-tag (vectors mentioned above). The immunoprecipitated complex can be examined for protein association by procedures such as Western blotting, SSS-methionine labeling of proteins, protein microsequencing, silver staining and two-dimensional gel electrophoresis.
[0206] Small molecules and ligands that interact with PSCA can be identified through related embodiments of such screening assays. For example, small molecules can be identified that interfere with protein function, including molecules that interfere with PSCA's ability to mediate phosphorylation and de-phosphorylation, interaction with DNA or RNA molecules as an indication of regulation of cell cycles, second messenger signaling or tumorigenesis. Similarly, small molecules that modulate PSCA-related ion channel, protein pump, or cell communication functions are identified and used WO 2005/014780 PCT/US2004/017231 to treat patients that have a cancer that expresses PSCA (see, Hille, Ionic Channels of Excitable Membranes 2 n d Ed., Sinauer Assoc., Sunderland, MA, 1992). Moreover, ligands that regulate PSCA function can be identified based on their ability to bind PSCA and activate a reporter construct. Typical methods are discussed for example in U.S. Patent No.
5,928,868 issued 27 July 1999, and include methods for forming hybrid ligands in which at least one ligand is a small molecule. In an illustrative embodiment, cells engineered to express a fusion protein of PSCA and a DNA-binding protein are used to co-express a fusion protein of a hybrid ligand/small molecule and a cDNA library transcriptional activator protein. The cells further contain a reporter gene, the expression of which is conditioned on the proximity of the first and second fusion proteins to each other, an event that occurs only if the hybrid ligand binds to target sites on both hybrid proteins. Those cells that express the reporter gene are selected and the unknown small molecule or the unknown ligand is identified. This method provides a means of identifying modulators, which activate or inhibit PSCA.
[0207] An embodiment of this invention comprises a method of screening for a molecule that interacts with a PSCA amino acid sequence shown in Figure 2 or Figure 3, comprising the steps of contacting a population of molecules with a PSCA amino acid sequence, allowing the population of molecules and the PSCA amino acid sequence to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the PSCA amino acid sequence, and then separating molecules that do not interact with the PSCA amino acid sequence from molecules that do.
In a specific embodiment, the method further comprises purifying, characterizing and identifying a molecule that interacts with the PSCA amino acid sequence. The identified molecule can be used to modulate a function performed by PSCA. In a preferred embodiment, the PSCA amino acid sequence is contacted with a library of peptides.
Therapeutic Methods and Compositions [0208] The identification of PSCA as a protein that is normally expressed in a restricted set of tissues, but which is also expressed in cancers such as those listed in Table I, opens a number of therapeutic approaches to the treatment of such cancers.
[0209] Of note, targeted antitumor therapies have been useful even when the targeted protein is expressed on normal tissues, even vital normal organ tissues. A vital organ is one that is necessary to sustain life, such as the heart or colon. A non-vital organ is one that can be removed whereupon the individual is still able to survive. Examples of non-vital organs are ovary, breast, and prostate.
[0210] For example, Herceptin® is an FDA approved pharmaceutical that has as its active ingredient an antibody which is immunoreactive with the protein variously known as HER2, HER2/neu, and erb-b-2. It is marketed by Genentech and has been a commercially successful antitumor agent. Herceptin sales reached almost $400 million in 2002. Herceptin is a treatment for HER2 positive metastatic breast cancer. However, the expression of HER2 is not limited to such tumors.
The same protein is expressed in a number of normal tissues. In particular, it is known that HER2/neu is present in normal kidney and heart, thus these tissues are present in all human recipients of Herceptin. The presence of HER2/neu in normal kidney is also confirmed by Latif, et al., B.J.U. International (2002) 89:5-9. As shown in this article (which evaluated whether renal cell carcinoma should be a preferred indication for anti-HER2 antibodies such as Herceptin) both protein and mRNA are produced in benign renal tissues. Notably, HER2/neu protein was strongly overexpressed in benign renal tissue.
[0211] Despite the fact that HER2/neu is expressed in such vital tissues as heart and kidney, Herceptin is a very useful, FDA approved, and commercially successful drug. The effect of Herceptin on cardiac tissue, "cardiotoxicity," WO 2005/014780 PCT/US2004/017231 has merely been a side effect to treatment. When patients were treated with Herceptin alone, significant cardiotoxicity occurred in a very low percentage of patients.
[0212] Of particular note, although kidney tissue is indicated to exhibit normal expression, possibly even higher expression than cardiac tissue, kidney has no appreciable Herceptin side effect whatsoever. Moreover, of the diverse array of normal tissues in which HER2 is expressed, there is very little occurrence of any side effect. Only cardiac tissue has manifested any appreciable side effect at all. A tissue such as kidney, where HER2/neu expression is especially notable, has not been the basis for any side effect.
[0213] Furthermore, favorable therapeutic effects have been found for antitumor therapies that target epidermal growth factor receptor (EGFR). EGFR is also expressed in numerous normal tissues. There have been very limited side effects in normal tissues following use of anti-EGFR therapeutics.
[0214] Thus, expression of a target protein in normal tissue, even vital normal tissue, does not defeat the utility of a targeting agent for the protein as a therapeutic for certain tumors in which the protein is also overexpressed.
[0215] Accordingly, therapeutic approaches that inhibit the activity of a PSCA protein are useful for patients suffering from a cancer that expresses PSCA. These therapeutic approaches generally fall into two classes. One class comprises various methods for inhibiting the binding or association of a PSCA protein with its binding partner or with other proteins.
Another class comprises a variety of methods for inhibiting the transcription of a PSCA gene or translation of PSCA mRNA.
Anti-Cancer Vaccines [0216] The invention provides cancer vaccines comprising a PSCA-related protein or PSCA-related nucleic acid. In view of the expression of PSCA, cancer vaccines prevent and/or treat PSCA-expressing cancers with minimal or no effects on non-target tissues. The use of a tumor antigen in a vaccine that generates humoral and/or cell-mediated immune responses as anti-cancer therapy is well known in the art and has been employed in prostate cancer using human PSMA and rodent PAP immunogens (Hodge et al, 1995, Int. J. Cancer 63:231-237; Fong et al., 1997, J. Immunol. 159:3113- 3117).
[0217] Such methods can be readily practiced by employing a PSCA-related protein, or a PSCA-encoding nucleic acid molecule and recombinant vectors capable of expressing and presenting the PSCA immunogen (which typically comprises a number of antibody or T cell epitopes). Skilled artisans understand that a wide variety of vaccine systems for delivery of immunoreactive epitopes are known in the art (see, Heryln et Ann Med 1999 Feb 31(1):66-78; Maruyama et al., Cancer Immunol Immunother 2000 Jun 49(3):123-32) Briefly, such methods of generating an immune response humoral and/or cell-mediated) in a mammal, comprise the steps of: exposing the mammal's immune system to an immunoreactive epitope an epitope present in a PSCA protein shown in Figure 3 or analog or homolog thereof so that the mammal generates an immune response that is specific for that epitope generates antibodies that specifically recognize that epitope). In a preferred method, a PSCA immunogen contains a biological motif, see e.g., Tables VIII-XXI and XXII-XLIX, or a peptide of a size range from PSCA indicated in Figure 5, Figure 6, Figure 7, Figure 8, and Figure 9.
[0218] The entire PSCA protein, immunogenic regions or epitopes thereof can be combined and delivered by various means. Such vaccine compositions can include, for example, lipopeptides (e.g.,Vitiello, A. et J. Clin. Invest.
95:341, 1995), peptide compositions encapsulated in poly(DL-lactide-co-glycolide) microspheres (see, e.g., Eldridge, et al., Molec. Immunol. 28:287-294, 1991: Alonso et Vaccine 12:299-306, 1994; Jones et Vaccine 13:675- 681, 1995), peptide compositions contained in immune stimulating complexes (ISCOMS) (see, Takahashi et a., 52 WO 2005/014780 PCT/US2004/017231 Nature 344:873-875, 1990; Hu et al, Clin Exp Immunol. 113:235-243, 1998), multiple antigen peptide systems (MAPs) (see Tam, J. Proc. Natl. Acad. Sci. U.S.A. 85:5409-5413, 1988; Tam, J. Immunol. Methods 196:17-32, 1996), peptides formulated as multivalent peptides; peptides for use in ballistic delivery systems, typically crystallized peptides, viral delivery vectors (Perkus, M. E. et al, In: Concepts in vaccine development, Kaufmann, S. H. ed., p. 379, 1996; Chakrabarti, S. et al., Nature 320:535, 1986; Hu, S. L. et al., Nature 320:537, 1986; Kieny, et al., AIDS Blo/Technology 4:790, 1986; Top, F. H. et J. Infect. Dis. 124:148,1971; Chanda, P. K. et al., Virology 175:535, 1990), particles of viral or synthetic origin Kofler, N. et J. Immunol. Methods. 192:25, 1996; Eldridge, J. H. et Sem.
Hematol. 30:16, 1993; Falo, L. Jr. et al., Nature Med. 7:649,1995), adjuvants (Warren, H. Vogel, F. and Chedid, L. A. Annu. Rev. Immunol. 4:369, 1986; Gupta, R. K. et Vaccine 11:293, 1993), liposomes (Reddy, R. et al., J.
Immunol. 148:1585, 1992; Rock, K. Immunol. Today 17:131, 1996), or, naked or particle absorbed cDNA (Ulmer, J. B. et al, Science 259:1745, 1993; Robinson, H. Hunt, L. and Webster, R. Vaccine 11:957, 1993; Shiver, J. W. etal., In: Concepts in vaccine development, Kaufmann, S. H. ed., p. 423, 1996; Cease, K. and Berzofsky, J. Annu.
Rev. Immunol. 12:923, 1994 and Eldridge, J. H. et Sem. Hematol. 30:16, 1993). Toxin-targeted delivery technologies, also known as receptor mediated targeting, such as those of Avant Immunotherapeutics, Inc. (Needham, Massachusetts) may also be used.
[0219] In patients with PSCA-associated cancer, the vaccine compositions of the invention can also be used in conjunction with other treatments used for cancer, surgery, chemotherapy, drug therapies, radiation therapies, etc.
including use in combination with immune adjuvants such as IL-2, IL-12, GM-CSF, and the like.
Cellular Vaccines: [0220] CTL epitopes can be determined using specific algorithms to identify peptides within PSCA protein that bind corresponding HLA alleles (see Table IV; Epimer T M and Epimatrix T M Brown University (URL brown.edu/Research/TB- HIV_Lab/epimatrixepimatrix.html); and, BIMAS, (URL bimas.dcrt.nih.gov/; SYFPEITHI at URL syfpeithi.bmiheidelberg.com/). In a preferred embodiment, a PSCA immunogen contains one or more amino acid sequences identified using techniques well known in the art, such as the sequences shown in Tables VIII-XXI and XXII-XLIX or a peptide of 8, 9, or 11 amino acids specified by an HLA Class I motif/supermotif Table IV Table IV or Table IV and/or a peptide of at least 9 amino acids that comprises an HLA Class II motif/supermotif Table IV or Table IV As is appreciated in the art, the HLA Class I binding groove is essentially closed ended so that peptides of only a particular size range can fit into the groove and be bound, generally HLA Class I epitopes are 8, 9, 10, or 11 amino acids long. In contrast, the HLA Class II binding groove is essentially open ended; therefore a peptide of about 9 or more amino acids can be bound by an HLA Class II molecule. Due to the binding groove differences between HLA Class I and II, HLA Class I motifs are length specific, position two of a Class I motif is the second amino acid in an amino to carboxyl direction of the peptide. The amino acid positions in a Class II motif are relative only to each other, not the overall peptide, i.e., additional amino acids can be attached to the amino and/or carboxyl termini of a motif-bearing sequence. HLA Class II epitopes are often 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acids long, or longer than amino acids.
WO 2005/014780 PCT/US2004/017231 Antibody-based Vaccines [0221] A wide variety of methods for generating an immune response in a mammal are known in the art (for example as the first step in the generation of hybridomas). Methods of generating an immune response in a mammal comprise exposing the mammal's immune system to an immunogenic epitope on a protein a PSCA protein) so that an immune response is generated. A typical embodiment consists of a method for generating an immune response to PSCA in a host, by contacting the host with a sufficient amount of at least one PSCA B cell or cytotoxic T-cell epitope or analog thereof; and at least one periodic interval thereafter re-contacting the host with the PSCA B cell or cytotoxic T-cell epitope or analog thereof. A specific embodiment consists of a method of generating an immune response against a PSCA-related protein or a man-made multiepitopic peptide comprising: administering PSCA immunogen a PSCA protein or a peptide fragment thereof, a PSCA fusion protein or analog etc.) in a vaccine preparation to a human or another mammal. Typically, such vaccine preparations further contain a suitable adjuvant (see, U.S. Patent No. 6,146,635) or a universal helper epitope such as a PADREM peptide (Epimmune Inc., San Diego, CA; see, Alexander ef al., J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et al, Immunity 1994 751-761 and Alexander et Immunol. Res. 1998 18(2): 79-92). An alternative method comprises generating an immune response in an individual against a PSCA immunogen by: administering in vivo to muscle or skin of the individual's body a DNA molecule that comprises a DNA sequence that encodes a PSCA immunogen, the DNA sequence operatively linked to regulatory sequences which control the expression of the DNA sequence; wherein the DNA molecule is taken up by cells, the DNA sequence is expressed in the cells and an immune response is generated against the immunogen (see, U.S. Patent No. 5,962,428). Optionally a genetic vaccine facilitator such as anionic lipids; saponins; lectins; estrogenic compounds; hydroxylated lower alkyls; dimethyl sulfoxide; and urea is also administered. In addition, an antiidiotypic antibody can be administered that mimics PSCA, in order to generate a response to the target antigen.
Nucleic Acid Vaccines: [0222] Vaccine compositions of the invention include nucleic acid-mediated modalities. DNA or RNA that encode protein(s) of the invention can be administered to a patient. Genetic immunization methods can be employed to generate prophylactic or therapeutic humoral and cellular immune responses directed against cancer cells expressing PSCA.
Constructs comprising DNA encoding a PSCA-related protein/immunogen and appropriate regulatory sequences can be injected directly into muscle or skin of an individual, such that the cells of the muscle or skin take-up the construct and express the encoded PSCA protein/immunogen. Alternatively, a vaccine comprises a PSCA-related protein. Expression of the PSCA-related protein immunogen results in the generation of prophylactic or therapeutic humoral and cellular immunity against cells that bear a PSCA protein. Various prophylactic and therapeutic genetic immunization techniques known in the art can be used (for review, see information and references published at Internet address genweb.com). Nucleic acidbased delivery is described, for instance, in Wolff et. al., Science 247:1465 (1990) as well as U.S. Patent Nos. 5,580,859; 5,589,466; 5,804,566; 5,739,118; 5,736,524; 5,679,647; WO 98/04720. Examples of DNA-based delivery technologies include "naked DNA", facilitated (bupivicaine, polymers, peptide-mediated) delivery, cationic lipid complexes, and particlemediated ("gene gun") or pressure-mediated delivery (see, U.S. Patent No. 5,922,687).
[0223] For therapeutic or prophylactic immunization purposes, proteins of the invention can be expressed via viral or bacterial vectors. Various viral gene delivery systems that can be used in the practice of the invention include, but are not limited to, vaccinia, fowlpox, canarypox, adenovirus, influenza, poliovirus, adeno-associated virus, lentivirus, and sindbis virus WO 2005/014780 PCT/US2004/017231 (see, Restifo, 1996, Curr. Opin. Immunol. 8:658-663; Tsang et al. J. Natl. Cancer Inst. 87:982-990 (1995)). Non-viral delivery systems can also be employed by introducing naked DNA encoding a PSCA-related protein into the patient intramuscularly or intradermally) to induce an anti-tumor response.
[0224] Vaccinia virus is used, for example, as a vector to express nucleotide sequences that encode the peptides of the invention. Upon introduction into a host, the recombinant vaccinia virus expresses the protein immunogenic peptide, and thereby elicits a host immune response. Vaccinia vectors and methods useful in immunization protocols are described in, U.S. Patent No. 4,722,848. Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover etal., Nature 351:456-460 (1991). A wide variety of other vectors useful for therapeutic administration or immunization of the peptides of the invention, e.g. adeno and adeno-associated virus vectors, retroviral vectors, Salmonella typhi vectors, detoxified anthrax toxin vectors, and the like, will be apparent to those skilled in the art from the description herein.
[0225] Thus, gene delivery systems are used to deliver a PSCA-related nucleic acid molecule. In one embodiment, the full-length human PSCA cDNA is employed. In another embodiment, PSCA nucleic acid molecules encoding specific cytotoxic T lymphocyte (CTL) and/or antibody epitopes are employed.
Ex Vivo Vaccines [0226] Various ex vive strategies can also be employed to generate an immune response. One approach involves the use of antigen presenting cells (APCs) such as dendritic cells (DC) to present PSCA antigen to a patient's immune system. Dendritic cells express MHC class I and II molecules, B7 co-stimulator, and IL-12, and are thus highly specialized antigen presenting cells. In prostate cancer, autologous dendritic cells pulsed with peptides of the prostate-specific membrane antigen (PSMA) are being used in a Phase I clinical trial to stimulate prostate cancer patients' immune systems (Tjoa et 1996, Prostate 28:65-69; Murphy et al., 1996, Prostate 29:371-380). Thus, dendritic cells can be used to present PSCA peptides to T cells in the context of MHC class I or II molecules. In one embodiment, autologous dendritic cells are pulsed with PSCA peptides capable of binding to MHC class I and/or class II molecules. In another embodiment, dendritic cells are pulsed with the complete PSCA protein. Yet another embodiment involves engineering the overexpression of a PSCA gene in dendritic cells using various implementing vectors known in the art, such as adenovirus (Arthur et al., 1997, Cancer Gene Ther. 4:17-25), retrovirus (Henderson et 1996, Cancer Res. 56:3763-3770), lentivirus, adeno-associated virus, DNA transfection (Ribas et al, 1997, Cancer Res. 57:2865-2869), or tumor-derived RNA transfection (Ashley et al., 1997, J. Exp. Med. 186:1177-1182). Cells that express PSCA can also be engineered to express immune modulators, such as GM-CSF, and used as immunizing agents.
PSCA as a Target for Antibody-based Therapy [0227] 511582008800able the use of reduced dosages of concomitant chemotherapy, particularly for patients who do not tolerate the toxicity of the chemotherapeutic agent very well.
[0228] Cancer patients can be evaluated for the presence and level of PSCA expression, preferably using immunohistochemical assessments of tumor tissue, quantitative PSCA imaging, or other techniques that reliably indicate the presence and degree of PSCA expression. Immunohistochemical analysis of tumor biopsies or surgical specimens is preferred for this purpose. Methods for immunohistochemical analysis of tumor tissues are well known in the art.
[0229] Anti-PSCA monoclonal antibodies that treat prostate and other cancers include those that initiate a potent immune response against the tumor or those that are directly cytotoxic. In this regard, anti-PSCA monoclonal antibodies .(mAbs) can elicit tumor cell lysis by either complement-mediated or antibody-dependent cell cytotoxicity (ADCC) WO 2005/014780 PCT/US2004/017231 mechanisms, both of which require an intact Fc portion of the immunoglobulin molecule for interaction with effector cell Fc receptor sites on complement proteins. In addition, anti-PSCA mAbs that exert a direct biological effect on tumor growth are useful to treat cancers that express PSCA. Mechanisms by which directly cytotoxic mAbs act include: inhibition of cell growth, modulation of cellular differentiation, modulation of tumor angiogenesis factor profiles, and the induction of apoptosis. The mechanism(s) by which a particular anti-PSCA mAb exerts an anti-tumor effect is evaluated using any number of in vitro assays that evaluate cell death such as ADCC, ADMMC, complement-mediated cell lysis, and so forth, as is generally known in the art.
[0230] In some patients, the use of murine or other non-human monoclonal antibodies, or human/mouse chimeric mAbs can induce moderate to strong immune responses against the non-human antibody. This can result in clearance of the antibody from circulation and reduced efficacy. In the most severe cases, such an immune response can lead to the extensive formation of immune complexes which, potentially, can cause renal failure. Accordingly, preferred monoclonal antibodies used in the therapeutic methods of the invention are those that are either fully human or humanized and that bind specifically to the target PSCA antigen with high affinity but exhibit low or no antigenicity in the patient.
[0231] Therapeutic methods of the invention contemplate the administration of single anti-PSCA mAbs as well as combinations, or cocktails, of different mAbs. Such mAb cocktails can have certain advantages inasmuch as they contain mAbs that target different epitopes, exploit different effector mechanisms or combine directly cytotoxic mAbs with mAbs that rely on immune effector functionality. Such mAbs in combination can exhibit synergistic therapeutic effects. In addition, anti-PSCA mAbs can be administered concomitantly with other therapeutic modalities, including but not limited to various chemotherapeutic agents, androgen-blockers, immune modulators IL-2, GM-CSF), surgery or radiation. The anti- PSCA mAbs are administered in their "naked" or unconjugated form, or can have a therapeutic agent(s) conjugated to them.
[0232] Anti-PSCA antibody formulations are administered via any route capable of delivering the antibodies to a tumor cell. Routes of administration include, but are not limited to, intravenous, intraperitoneal, intramuscular, intratumor, intradermal, and the like. Treatment generally involves repeated administration of the anti-PSCA antibody preparation, via an acceptable route of administration such as intravenous injection typically at a dose in the range of about 0.1, .3, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 mg/kg body weight. In general, doses in the range of 10-1000 mg mAb per week are effective and well tolerated.
[0233] Based on clinical experience with the HerceptinTM mAb in the treatment of metastatic breast cancer, an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti- PSCA mAb preparation represents an acceptable dosing regimen. Preferably, the initial loading dose is administered as a or longer infusion. The periodic maintenance dose is administered as a 30 minute or longer infusion, provided the initial dose was well tolerated. As appreciated by those of skill in the art, various factors can influence the ideal dose regimen in a particular case. Such factors include, for example, the binding affinity and half life of the Ab or mAbs used, the degree of PSCA expression in the patient, the extent of circulating shed PSCA antigen, the desired steady-state antibody concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient.
[0234] Optionally, patients should be evaluated for the levels of PSCA in a given sample the levels of circulating PSCA antigen and/or PSCA expressing cells) in order to assist in the determination of the most effective dosing regimen, etc. Such evaluations are also used for monitoring purposes throughout therapy, and are useful to gauge WO 2005/014780 PCT/US2004/017231 therapeutic success in combination with the evaluation of other parameters (for example, urine cytology and/or ImmunoCyt levels in bladder cancer therapy, or by analogy, serum PSA levels in prostate cancer therapy).
[0235] Anti-idiotypic anti-PSCA antibodies can also be used in anti-cancer therapy as a vaccine for inducing an immune response to cells expressing a PSCA-related protein. In particular, the generation of anti-idiotypic antibodies is well known in the art; this methodology can readily be adapted to generate anti-idiotypic anti-PSCA antibodies that mimic an epitope on a PSCA-related protein (see, for example, Wagner etal., 1997, Hybridoma 16: 33-40; Foon et al., 1995, J.
Clin. Invest. 96:334-342; Herlyn et al., 1996, Cancer immunol. Immunother. 43:65-76). Such an anti-idiotypic antibody can be used in cancer vaccine strategies.
PSCA as a Taraet for Cellular Immune Responses [0236] Vaccines and methods of preparing vaccines that contain an immunogenically effective amount of one or more HLA-binding peptides as described herein are further embodiments of the invention. Furthermore, vaccines in accordance with the invention encompass compositions of one or more of the claimed peptides. A peptide can be present in a vaccine individually. Alternatively, the peptide can exist as a homopolymer comprising multiple copies of the same peptide, or as a heteropolymer of various peptides. Polymers have the advantage of increased immunological reaction and, where different peptide epitopes are used to make up the polymer, the additional ability to induce antibodies and/or CTLs that react with different antigenic determinants of the pathogenic organism or tumor-related peptide targeted for an immune response. The composition can be a naturally occurring region of an antigen or can be prepared, e.g., recombinantly or by chemical synthesis.
[0237] Carriers that can be used with vaccines of the invention are well known in the art, and include, e.g., thyroglobulin, albumins such as human serum albumin, tetanus toxoid, polyamino acids such as poly L-lysine, poly Lglutamic acid, influenza, hepatitis B virus core protein, and the like. The vaccines can contain a physiologically tolerable acceptable) diluent such as water, or saline, preferably phosphate buffered saline. The vaccines also typically include an adjuvant. Adjuvants such as incomplete Freund's adjuvant, aluminum phosphate, aluminum hydroxide, or alum are examples of materials well known in the art. Additionally, as disclosed herein, CTL responses can be primed by conjugating peptides of the invention to lipids, such as tripalmitoyl-S-glycerylcysteinlyseryl- serine (P 3 CSS). Moreover, an adjuvant such as a synthetic cytosine-phosphorothiolated-guanine-containing (CpG) oligonucleotides has been found to increase CTL responses 10- to 100-fold. (see, e.g. Davila and Cells, J. Immunol. 165:539-547 (2000)) [0238] Upon immunization with a peptide composition in accordance with the invention, via injection, aerosol, oral, transdermal, transmucosal, intrapleural, intrathecal, or other suitable routes, the immune system of the host responds to the vaccine by producing large amounts of CTLs and/or HTLs specific for the desired antigen. Consequently, the host becomes at least partially immune to later development of cells that express or overexpress PSCA antigen, or derives at least some therapeutic benefit when the antigen was tumor-associated.
[0239] In some embodiments, it may be desirable to combine the class I peptide components with components that induce or facilitate neutralizing antibody and or helper T cell responses directed to the target antigen. A preferred embodiment of such a composition comprises class I and class II epitopes in accordance with the invention. An alternative embodiment of such a composition comprises a class I and/or class II epitope in accordance with the invention, along with a cross reactive HTL epitope such as PADRETM (Epimmune, San Diego, CA) molecule (described in U.S. Patent Number 5,736,142).
WO 2005/014780 PCT/US2004/017231 [0240] A vaccine of the invention can also include antigen-presenting cells (APC), such as dendritic cells as a vehicle to present peptides of the invention. Vaccine compositions can be created in vitro, following dendritic cell mobilization and harvesting, whereby loading of dendritic cells occurs in vitro. For example, dendritic cells are transfected, with a minigene in accordance with the invention, or are pulsed with peptides. The dendritic cell can then be administered to a patient to elicit immune responses in vivo. Vaccine compositions, either DNA- or peptide-based, can also be administered in vivo in combination with dendritic cell mobilization whereby loading of dendritic cells occurs in vivo.
[0241] Preferably, the following principles are utilized when selecting an array of epitopes for inclusion in a polyepitopic composition for use in a vaccine, or for selecting discrete epitopes to be included in a vaccine and/or to be encoded by nucleic acids such as a minigene. It is preferred that each of the following principles be balanced in order to make the selection. The multiple epitopes to be incorporated in a given vaccine composition may be, but need not be, contiguous in sequence in the native antigen from which the epitopes are derived.
Epitopes are selected which, upon administration, mimic immune responses that have been observed to be correlated with tumor clearance, For HLA Class I this includes 3-4 epitopes that come from at least one tumor associated antigen (TAA). For HLA Class II a similar rationale is employed; again 3-4 epitopes are selected from at least one TAA (see, Rosenberg et al., Science 278:1447-1450). Epitopes from one TAA may be used in combination with epitopes from one or more additional TAAs to produce a vaccine that targets tumors with varying expression patterns of frequently-expressed TAAs.
Epitopes are selected that have the requisite binding affinity established to be correlated with immunogenicity: for HLA Class I an IC50 of 500 nM or less, often 200 nM or less; and for Class II an ICso of 1000 nM or less.
Sufficient supermotif bearing-peptides, or a sufficient array of allele-specific motif-bearing peptides, are selected to give broad population coverage. For example, it is preferable to have at least 80% population coverage. A Monte Carlo analysis, a statistical evaluation known in the art, can be employed to assess the breadth, or redundancy of, population coverage.
When selecting epitopes from cancer-related antigens it is often useful to select analogs because the patient may have developed tolerance to the native epitope.
Of particular relevance are epitopes referred to as "nested epitopes." Nested epitopes occur where at least two epitopes overlap in a given peptide sequence. A nested peptide sequence can comprise B cell, HLA class I and/or HLA class II epitopes. When providing nested epitopes, a general objective is to provide the greatest number of epitopes per sequence. Thus, an aspect is to avoid providing a peptide that is any longer than the amino terminus of the amino terminal epitope and the carboxyl terminus of the carboxyl terminal epitope in the peptide. When providing a multiepitopic sequence, such as a sequence comprising nested epitopes, it is generally important to screen the sequence in order to insure that it does not have pathological or other deleterious biological properties.
If a polyepitopic protein is created, or when creating a minigene, an objective is to generate the smallest peptide that encompasses the epitopes of interest. This principle is similar, if not the same as that employed when selecting a peptide comprising nested epitopes. However, with an artificial polyepitopic peptide, the size minimization objective is balanced against the need to integrate any spacer sequences between epitopes in the polyepitopic protein.
Spacer amino acid residues can, for example, be introduced to avoid junctional epitopes (an epitope recognized by the immune system, not present in the target antigen, and only created by the man-made juxtaposition of epitopes), or to facilitate cleavage between epitopes and thereby enhance epitope presentation. Junctional epitopes are generally to be 58 WO 2005/014780 PCT/US2004/017231 avoided because the recipient may generate an immune response to that non-native epitope. Of particular concern is a junctional epitope that is a "dominant epitope." A dominant epitope may lead to such a zealous response that immune responses to other epitopes are diminished or suppressed.
Where the sequences of multiple variants of the same target protein are present, potential peptide epitopes can also be selected on the basis of their conservancy. For example, a criterion for conservancy may define that the entire sequence of an HLA class I binding peptide or the entire 9-mer core of a class II binding peptide be conserved in a designated percentage of the sequences evaluated for a specific protein antigen.
X.C.1. Minigene Vaccines [0242] A number of different approaches are available which allow simultaneous delivery of multiple epitopes.
Nucleic acids encoding the peptides of the invention are a particularly useful embodiment of the invention. Epitopes for inclusion in a minigene are preferably selected according to the guidelines set forth in the previous section. A preferred means of administering nucleic acids encoding the peptides of the invention uses minigene constructs encoding a peptide comprising one or multiple epitopes of the invention.
[0243] The use of multi-epitope minigenes is described below and in, Ishioka et al., J. Immunol. 162:3915-3925, 1999; An, L. and Whitton, J. J. Viral 71:2292, 1997; Thomson, S. A. et J. Immunol. 157:822, 1996; Whitton, J. L. et al., J. Virol. 67:348, 1993; Hanke, R. et al., Vaccine 16:426, 1998. For example, a multi-epitope DNA plasmid encoding supermotif- andlor motif-bearing epitopes derived PSCA, the PADRE® universal helper T cell epitope or multiple HTL epitopes from PSCA (see Tables VIII-XXI and XXII to XLIX), and an endoplasmic reticulum-translocating signal sequence can be engineered. A vaccine may also comprise epitopes that are derived from other TAAs.
[0244] The immunogenicity of a multi-epitopic minigene can be confirmed in transgenic mice to evaluate the magnitude of CTL induction responses against the epitopes tested. Further, the immunogenicity of DNA-encoded epitopes in vivo can be correlated with the in vitro responses of specific CTL lines against target cells transfected with the DNA plasmid. Thus, these experiments can show that the minigene serves to both: generate a CTL response and that the induced CTLs recognized cells expressing the encoded epitopes.
[0245] For example, to create a DNA sequence encoding the selected epitopes (minigene) for expression in human cells, the amino acid sequences of the epitopes may be reverse translated. A human codon usage table can be used to guide the codon choice for each amino acid. These epitope-encoding DNA sequences may be directly adjoined, so that when translated, a continuous polypeptide sequence is created. To optimize expression and/or immunogenicity, additional elements can be incorporated into the minigene design. Examples of amino acid sequences that can be reverse translated and included in the minigene sequence include: HLA class I epitopes, HLA class II epitopes, antibody epitopes, a ubiquitination signal sequence, and/or an endoplasmic reticulum targeting signal. In addition, HLA presentation of CTL and HTL epitopes may be improved by including synthetic poly-alanine) or naturally-occurring flanking sequences adjacent to the CTL or HTL epitopes; these larger peptides comprising the epitope(s) are within the scope of the invention.
[0246] The minigene sequence may be converted to DNA by assembling oligonucleotides that encode the plus and minus strands of the minigene. Overlapping oligonucleotides (30-100 bases long) may be synthesized, phosphorylated, purified and annealed under appropriate conditions using well known techniques. The ends of the oligonucleotides can be joined, for example, using T4 DNA ligase. This synthetic minigene, encoding the epitope polypeptide, can then be cloned into a desired expression vector.
WO 2005/014780 PCT/US2004/017231 [0247] Standard regulatory sequences well known to those of skill in the art are preferably included in the vector to ensure expression in the target cells. Several vector elements are desirable: a promoter with a down-stream cloning site for minigene insertion; a polyadenylation signal for efficient transcription termination; an E. coli origin of replication; and an E. coliselectable marker ampicillin or kanamycin resistance). Numerous promoters can be used for this purpose, e.g., the human cytomegalovirus (hCMV) promoter. See, U.S. Patent Nos. 5,580,859 and 5,589,466 for other suitable promoter sequences.
[0248] Additional vector modifications may be desired to optimize minigene expression and immunogenicity. In some cases, introns are required for efficient gene expression, and one or more synthetic or naturally-occurring introns could be incorporated into the transcribed region of the minigene. The inclusion of mRNA stabilization sequences and sequences for replication in mammalian cells may also be considered for increasing minigene expression.
[0249] Once an expression vector is selected, the minigene is cloned into the polylinker region downstream of the promoter. This plasmid is transformed into an appropriate E. colistrain, and DNA is prepared using standard techniques.
The orientation and DNA sequence of the minigene, as well as all other elements included in the vector, are confirmed using restriction mapping and DNA sequence analysis. Bacterial cells harboring the correct plasmid can be stored as a master cell bank and a working cell bank.
[0250] In addition, immunostimulatory sequences (ISSs or CpGs) appear to play a role in the immunogenicity of DNA vaccines. These sequences may be included in the vector, outside the minigene coding sequence, if desired to enhance immunogenicity.
[0251] In some embodiments, a bi-cistronic expression vector which allows production of both the minigene-encoded epitopes and a second protein (included to enhance or decrease immunogenicity) can be used. Examples of proteins or polypeptides that could beneficially enhance the immune response if co-expressed include cytokines IL-2, IL-12, GM- CSF), cytokine-inducing molecules LelF), costimulatory molecules, or for HTL responses, pan-DR binding proteins
(PADRE
T M Epimmune, San Diego, CA). Helper (HTL) epitopes can be joined to intracellular targeting signals and expressed separately from expressed CTL epitopes; this allows direction of the HTL epitopes to a cell compartment different than that of the CTL epitopes. If required, this could facilitate more efficient entry of HTL epitopes into the HLA class II pathway, thereby improving HTL induction. In contrast to HTL or CTL induction, specifically decreasing the immune response by co-expression of immunosuppressive molecules TGF-3) may be beneficial in certain diseases.
[0252] Therapeutic quantities of plasmid DNA can be produced for example, by fermentation in E. coli, followed by purification. Aliquots from the working cell bank are used to inoculate growth medium, and grown to saturation in shaker flasks or a bioreactor according to well-known techniques. Plasmid DNA can be purified using standard bioseparation technologies such as solid phase anion-exchange resins supplied by QIAGEN, Inc. (Valencia, California). If required, supercoiled DNA can be isolated from the open circular and linear forms using gel electrophoresis or other methods.
[0253] Purified plasmid DNA can be prepared for injection using a variety of formulations. The simplest of these is reconstitution of lyophilized DNA in sterile phosphate-buffer saline (PBS). This approach, known as "naked DNA," is currently being used for intramuscular (IM) administration in clinical trials. To maximize the immunotherapeutic effects of minigene DNA vaccines, an alternative method for formulating purified plasmid DNA may be desirable. A variety of methods have been described, and new techniques may become available. Cationic lipids, glycolipids, and fusogenic liposomes can also be used in the formulation (see, as described by WO 93/24640; Mannino Gould-Fogerite, BioTechniques 682 (1988); U.S. Pat No. 5,279,833; WO 91/06309; and Feigner, et al., Proc. Nat'l Acad. Sci. USA WO 2005/014780 PCT/US2004/017231 84:7413 (1987). In addition, peptides and compounds referred to collectively as protective, interactive, non-condensing compounds (PINC) could also be complexed to purified plasmid DNA to influence variables such as stability, intramuscular dispersion, or trafficking to specific organs or cell types.
[0254] Target cell sensitization can be used as a functional assay for expression and HLA class I presentation of minigene-encoded CTL epitopes. For example, the plasmid DNA is introduced into a mammalian cell line that is suitable as a target for standard CTL chromium release assays. The transfection method used will be dependent on the final formulation. Electroporation can be used for "naked" DNA, whereas cationic lipids allow direct in vitro transfection. A plasmid expressing green fluorescent protein (GFP) can be co-transfected to allow enrichment of transfected cells using fluorescence activated cell sorting (FACS). These cells are then chromium-51 5 1Cr) labeled and used as target cells for epitope-specific CTL lines; cytolysis, detected by 51 Cr release, indicates both production of, and HLA presentation of, minigene-encoded CTL epitopes. Expression of HTL epitopes may be evaluated in an analogous manner using assays to assess HTL activity.
[0255] In vivo immunogenicity is a second approach for functional testing of minigene DNA formulations. Transgenic mice expressing appropriate human HLA proteins are immunized with the DNA product. The dose and route of administration are formulation dependent IM for DNA in PBS, intraperitoneal for lipid-complexed DNA). Twentyone days after immunization, splenocytes are harvested and restimulated for one week in the presence of peptides encoding each epitope being tested. Thereafter, for CTL effector cells, assays are conducted for cytolysis of peptideloaded, 51 Cr-labeled target cells using standard techniques. Lysis of target cells that were sensitized by HLA loaded with peptide epitopes, corresponding to minigene-encoded epitopes, demonstrates DNA vaccine function for in vivo induction of CTLs. Immunogenicity of HTL epitopes is confirmed in transgenic mice in an analogous manner.
[0256] Alternatively, the nucleic acids can be administered using ballistic delivery as described, for instance, in U.S.
Patent No. 5,204,253. Using this technique, particles comprised solely of DNA are administered. In a further alternative embodiment, DNA can be adhered to particles, such as gold particles.
[0257] Minigenes can also be delivered using other bacterial or viral delivery systems well known in the art, an expression construct encoding epitopes of the invention can be incorporated into a viral vector such as vaccinia.
X.C.2. Combinations of CTL Peptides with Helper Peptides [0258] Vaccine compositions comprising CTL peptides of the invention can be modified, analoged, to provide desired attributes, such as improved serum half life, broadened population coverage or enhanced immunogenicity.
[0259] For instance, the ability of a peptide to induce CTL activity can be enhanced by linking the peptide to a sequence which contains at least one epitope that is capable of inducing a T helper cell response. Although a CTL peptide can be directly linked to a T helper peptide, often CTL epitope/HTL epitope conjugates are linked by a spacer molecule.
The spacer is typically comprised of relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions. The spacers are typically selected from, Ala, Gly, or other neutral spacers of nonpolar amino acids or neutral polar amino acids. It will be understood that the optionally present spacer need not be comprised of the same residues and thus may be a hetero- or homo-oligomer. When present, the spacer will usually be at least one or two residues, more usually three to six residues and sometimes 10 or more residues.
The CTL peptide epitope can be linked to the T helper peptide epitope either directly or via a spacer either at the amino or carboxy terminus of the CTL peptide. The amino terminus of either the immunogenic peptide or the T helper peptide may be acylated.
WO 2005/014780 PCT/US2004/017231 [0260] In certain embodiments, the T helper peptide is one that is recognized by T helper cells present in a majority of a genetically diverse population. This can be accomplished by selecting peptides that bind to many, most, or all of the HLA class II molecules. Examples of such amino acid bind many HLA Class II molecules include sequences from antigens such as tetanus toxoidat positions 830-843 (QYIKANSKFIGITE; SEQ ID NO:1), Plasmodium falciparum circumsporozoite (CS) protein at positions 378-398 (DIEKKIAKMEKASSVFNVVNS; SEQ ID NO:2), and Streptococcus 18kD protein at positions 116-131 (GAVDSILGGVATYGAA; SEQ ID NO:3). Other examples include peptides bearing a DR 1-4-7 supermotif, or either of the DR3 motifs.
[0261] Alternatively, it is possible to prepare synthetic peptides capable of stimulating T helper lymphocytes, in a loosely HLA-restricted fashion, using amino acid sequences not found in nature (see, PCT publication WO 95/07707).
These synthetic compounds called Pan-DR-binding epitopes PADRETM, Epimmune, Inc., San Diego, CA) are designed, most preferably, to bind most HLA-DR (human HLA class II) molecules. For instance, a pan-DR-binding epitope peptide having the formula: aKXVAAWTLKAa (SEQ ID NO:4), where is either cyclohexylalanine, phenylalanine, or tyrosine, and a is either D-alanine or L-alanine, has been found to bind to most HLA-DR alleles, and to stimulate the response of T helper lymphocytes from most individuals, regardless of their HLA type. An alternative of a pan-DR binding epitope comprises all L" natural amino acids and can be provided in the form of nucleic acids that encode the epitope.
[0262] HTL peptide epitopes can also be modified to alter their biological properties. For example, they can be modified to include D-amino acids to increase their resistance to proteases and thus extend their serum half life, or they can be conjugated to other molecules such as lipids, proteins, carbohydrates, and the like to increase their biological activity.
For example, a T helper peptide can be conjugated to one or more palmitic acid chains at either the amino or carboxyl termini.
X.C.3. Combinations of CTL Peptides with T Cell Priming Agents [0263] In some embodiments it may be desirable to include in the pharmaceutical compositions of the invention at least one component which primes B lymphocytes or T lymphocytes. Lipids have been identified as agents capable of priming CTL in vivo. For example, palmitic acid residues can be attached to the s-and a- amino groups of a lysine residue and then linked, via one or more linking residues such as Gly, Gly-Gly-, Ser, Ser-Ser, or the like, to an immunogenic peptide. The lipidated peptide can then be administered either directly in a micelle or particle, incorporated into a liposome, or emulsified in an adjuvant, incomplete Freund's adjuvant. In a preferred embodiment, a particularly effective immunogenic composition comprises palmitic acid attached to e- and a- amino groups of Lys, which is attached via linkage, Ser-Ser, to the amino terminus of the immunogenic peptide.
[0264] As another example of lipid priming of CTL responses, E. coli lipoproteins, such as tripalmitoyl-Sglycerylcysteinlyseryl- serine (P3CSS) can be used to prime virus specific CTL when covalently attached to an appropriate peptide (see, Deres, etal., Nature 342:561, 1989). Peptides of the invention can be coupled to P3CSS, for example, and the lipopeptide administered to an individual to prime specifically an immune response to the target antigen. Moreover, because the induction of neutralizing antibodies can also be primed with PaCSS-conjugated epitopes, two such compositions can be combined to more effectively elicit both humoral and cell-mediated responses.
WO 2005/014780 PCT/US2004/017231 X.C.4. Vaccine Compositions Comprising DC Pulsed with CTL and/or HTL Peptides [0265] An embodiment of a vaccine composition in accordance with the invention comprises ex vivo administration of a cocktail of epitope-bearing peptides to PBMC, or isolated DC therefrom, from the patient's blood. A pharmaceutical to facilitate harvesting of DC can be used, such as Progenipoietin T m (Pharmacia-Monsanto, St. Louis, MO) or GM-CSF/IL-4.
After pulsing the DC with peptides and prior to reinfusion into patients, the DC are washed to remove unbound peptides. In this embodiment, a vaccine comprises peptide-pulsed DCs which present the pulsed peptide epitopes complexed with HLA molecules on their surfaces.
[0266] The DC can be pulsed ex vivo with a cocktail of peptides, some of which stimulate CTL responses to PSCA.
Optionally, a helper T cell (HTL) peptide, such as a natural or artificial loosely restricted HLA Class II peptide, can be included to facilitate the CTL response. Thus, a vaccine in accordance with the invention is used to treat a cancer which expresses or overexpresses PSCA.
X.D. Adoptive Immunotherapv [0267] Antigenic PSCA-related peptides are used to elicit a CTL and/or HTL response ex vivo, as well. The resulting CTL or HTL cells, can be used to treat tumors in patients that do not respond to other conventional forms of therapy, or will not respond to a therapeutic vaccine peptide or nucleic acid in accordance with the invention. Ex vivo CTL or HTL responses to a particular antigen are induced by incubating in tissue culture the patients, or genetically compatible, CTL or HTL precursor cells together with a source of antigen-presenting cells (APC), such as dendritic cells, and the appropriate immunogenic peptide. After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused back into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cell a tumor cell). Transfected dendritic cells may also be used as antigen presenting cells.
X.E. Administration of Vaccines for Therapeutic or Prophylactic Purposes [0268] Pharmaceutical and vaccine compositions of the invention are typically used to treat and/or prevent a cancer that expresses or overexpresses PSCA. In therapeutic applications, peptide and/or nucleic acid compositions are administered to a patient in an amount sufficient to elicit an effective B cell, CTL and/or HTL response to the antigen and to cure or at least partially arrest or slow symptoms and/or complications. An amount adequate to accomplish this is defined as "therapeutically effective dose." Amounts effective for this use will depend on, the particular composition administered, the manner of administration, the stage and severity of the disease being treated, the weight and general state of health of the patient, and the judgment of the prescribing physician.
[0269] For pharmaceutical compositions, the immunogenic peptides of the invention, or DNA encoding them, are generally administered to an individual already bearing a tumor that expresses PSCA. The peptides or DNA encoding them can be administered individually or as fusions of one or more peptide sequences. Patients can be treated with the immunogenic peptides separately or in conjunction with other treatments, such as surgery, as appropriate.
[0270] For therapeutic use, administration should generally begin at the first diagnosis of PSCA-associated cancer.
This is followed by boosting doses until at least symptoms are substantially abated and for a period thereafter. The embodiment of the vaccine composition including, but not limited to embodiments such as peptide cocktails, polyepitopic polypeptides, minigenes, or TAA-specific CTLs or pulsed dendritic cells) delivered to the patient may vary WO 2005/014780 PCT/US2004/017231 according to the stage of the disease or the patient's health status. For example, in a patient with a tumor that expresses PSCA, a vaccine comprising PSCA-specific CTL may be more efficacious in killing tumor cells in patient with advanced disease than alternative embodiments.
[0271] It is generally important to provide an amount of the peptide epitope delivered by a mode of administration sufficient to stimulate effectively a cytotoxic T cell response; compositions which stimulate helper T cell responses can also be given in accordance with this embodiment of the invention.
[0272] The dosage for an initial therapeutic immunization generally occurs in a unit dosage range where the lower value is about 1, 5, 50, 500, or 1,000 pg and the higher value is about 10,000; 20,000; 30,000; or 50,000 pg. Dosage values for a human typically range from about 500 pg to about 50,000 pg per 70 kilogram patient. Boosting dosages of between about 1.0 Ipg to about 50,000 pg of peptide pursuant to a boosting regimen over weeks to months may be administered depending upon the patient's response and condition as determined by measuring the specific activity of CTL and HTL obtained from the patient's blood. Administration should continue until at least clinical symptoms or laboratory tests indicate that the neoplasia, has been eliminated or reduced and for a period thereafter. The dosages, routes of administration, and dose schedules are adjusted in accordance with methodologies known in the art.
[0273] In certain embodiments, the peptides and compositions of the present invention are employed in serious disease states, that is, life-threatening or potentially life threatening situations. In such cases, as a result of the minimal amounts of extraneous substances and the relative nontoxic nature of the peptides in preferred compositions of the invention, it is possible and may be felt desirable by the treating physician to administer substantial excesses of these peptide compositions relative to these stated dosage amounts.
[0274] The vaccine compositions of the invention can also be used purely as prophylactic agents. Generally the dosage for an initial prophylactic immunization generally occurs in a unit dosage range where the lower value is about 1, 500, or 1000 pg and the higher value is about 10,000; 20,000; 30,000; or 50,000 pg. Dosage values for a human typically range from about 500 pg to about 50,000 pg per 70 kilogram patient. This is followed by boosting dosages of between about 1.0 pg to about 50,000 pg of peptide administered at defined intervals from about four weeks to six months after the initial administration of vaccine. The immunogenicity of the vaccine can be assessed by measuring the specific activity of CTL and HTL obtained from a sample of the patient's blood.
[0275] The pharmaceutical compositions for therapeutic treatment are Intended for parenteral, topical, oral, nasal, intrathecal, or local as a cream or topical ointment) administration. Preferably, the pharmaceutical compositions are administered parentally, intravenously, subcutaneously, intradermally, or intramuscularly. Thus, the invention provides compositions for parenteral administration which comprise a solution of the immunogenic peptides dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.
[0276] A variety of aqueous carriers may, be used, water, buffered water, 0.8% saline, 0.3% glycine, hyaluronic acid and the like. These compositions may be sterilized by conventional, well-known sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
[0277] The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservatives, and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
WO 2005/014780 PCT/US2004/017231 [0278] The concentration of peptides of the invention in the pharmaceutical formulations can vary widely, from less than about usually at or at least about 2% to as much as 20% to 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
[0279] A human unit dose form of a composition is typically included in a pharmaceutical composition that comprises a human unit dose of an acceptable carrier, in one embodiment an aqueous carrier, and is administered in a volume/quantity that is known by those of skill in the art to be used for administration of such compositions to humans (see, Remington's Pharmaceutical Sciences, 17 t Edition, A. Gennaro, Editor, Mack Publishing Co., Easton, Pennsylvania, 1985). For example a peptide dose for initial immunization can be from about 1 to about 50,000 pg, generally 100-5,000 pg, for a 70 kg patient. For example, for nucleic acids an initial immunization may be performed using an expression vector in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites. The nucleic acid (0.1 to 1000 pg) can also be administered using a gene gun. Following an incubation period of 3-4 weeks, a booster dose is then administered. The booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5x10 9 pfu.
[0280] For antibodies, a treatment generally involves repeated administration of the anti-PSCA antibody preparation, via an acceptable route of administration such as intravenous injection typically at a dose in the range of about 0.1 to about 10 mg/kg body weight. In general, doses in the range of 10-500 mg mAb per week are effective and well tolerated.
Moreover, an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti- PSCA mAb preparation represents an acceptable dosing regimen. As appreciated by those of skill in the art, various factors can influence the ideal dose in a particular case. Such factors include, for example, half life of a composition, the binding affinity of an Ab, the immunogenicity of a substance, the degree of PSCA expression in the patient, the extent of circulating shed PSCA antigen, the desired steady-state concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient. Non-limiting preferred human unit doses are, for example, 500pg 1mg, 1mg 50mg, 50mg 100mg, 100mg 200mg, 200mg 300mg, 400mg 500mg, 500mg 600mg, 600mg 700mg, 700mg 800mg, 800mg 900mg, 900mg 1g, or 1mg 700mg. In certain embodiments, the dose is in a range of 2-5 mg/kg body weight, with follow on weekly doses of 1-3 mg/kg; 0.5mg, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10mg/kg body weight followed, in two, three or four weeks by weekly doses; 0.5 10mg/kg body weight, followed in two, three or four weeks by weekly doses; 225, 250, 275, 300, 325, 350, 375, 400mg m 2 of body area weekly; 1-600mg m 2 of body area weekly; 225-400mg m 2 of body area weekly; these does can be followed by weekly doses for 2, 3, 4, 5, 6, 7, 8, 9, 19, 11, 12 or more weeks.
[0281] In one embodiment, human unit dose forms of polynucleotides comprise a suitable dosage range or effective amount that provides any therapeutic effect As appreciated by one of ordinary skill in the art a therapeutic effect depends on a number of factors, including the sequence of the polynucleotide, molecular weight of the polynucleotide and route of administration. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like. Generally, for a polynucleotide of about 20 bases, a dosage range may be selected from, for example, an independently selected lower limit such as about 0.1, 0.25, 0.5, 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400 or 500 mg/kg up to an independently selected upper limit, greater than the lower limit, of about 60, 80, 100, 200, 300, 400, 500, 750, 1000, 1500, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 or 10,000 mg/kg. For example, a dose may be about any of the following: 0.1 to 100 mg/kg, 0.1 to 50 mg/kg, 0.1 to 25 mg/kg, 0.1 to 10 mg/kg, 1 to 500 mg/kg, 100 to 400 mg/kg, 200 to 300 mg/kg, 1 to 100 mg/kg, 100 to 200 mg/kg, 300 to 400 mg/kg, 400 to 500 mg/kg, 500 to 1000 mg/kg, 500 to 5000 mg/kg, or 500 to WO 2005/014780 PCT/US2004/017231 10,000 mg/kg. Generally, parenteral routes of administration may require higher doses of polynucleotide compared to more direct application to the nucleotide to diseased tissue, as do polynucleotides of increasing length.
[0282] In one embodiment, human unit dose forms of T-cells comprise a suitable dosage range or effective amount that provides any therapeutic effect. As appreciated by one of ordinary skill in the art, a therapeutic effect depends on a number of factors. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like. A dose may be about 104 cells to about 106 cells, about 106 cells to about 108 cells, about 108 to about 1011 cells, or about 10 B to about 5 x 1010 cells. A dose may also about 106 cells/m 2 to about 1010 cells/m 2 or about 106 cells/m 2 to about 108 cells/m 2 [0283] Proteins(s) of the invention, and/or nucleic acids encoding the protein(s), can also be administered via liposomes, which may also serve to: 1) target the proteins(s) to a particular tissue, such as lymphoid tissue; 2) to target selectively to diseases cells; or, 3) to increase the half-life of the peptide composition. Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. In these preparations, the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the CD45 antigen, or with other therapeutic or immunogenic compositions. Thus, liposomes either filled or decorated with a desired peptide of the invention can be directed to the site of lymphoid cells, where the liposomes then deliver the peptide compositions.
Liposomes for use in accordance with the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, liposome size, acid lability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, Szoka, et al, Ann. Rev. Biophys. Bioeng. 9:467 (1980), and U.S. Patent Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369.
[0284] For targeting cells of the immune system, a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells. A liposome suspension containing a peptide may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the peptide being delivered, and the stage of the disease being treated.
[0285] For solid compositions, conventional nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. For oral administration, a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 10-95% of active ingredient, that is, one or more peptides of the invention, and more preferably at a concentration of [0286] For aerosol administration, immunogenic peptides are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of peptides are about 0.01%-20% by weight, preferably about 1%-10%.
The surfactant must, of course, be nontoxic, and preferably soluble in the propellant. Representative of such agents are the esters or partial esters of fatty acids containing from about 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride.
Mixed esters, such as mixed or natural glycerides may be employed. The surfactant may constitute about 0.1%-20% by weight of the composition, preferably about 0.25-5%. The balance of the composition is ordinarily propellant. A carrier can also be included, as desired, as with, lecithin for intranasal delivery.
WO 2005/014780 PCT/US2004/017231 XI.) Diagnostic and Prognostic Embodiments of PSCA.
[0287] As disclosed herein, PSCA polynucleotides, polypeptides, reactive cytotoxic T cells (CTL), reactive helper T cells (HTL) and anti-polypeptide antibodies are used in well known diagnostic, prognostic and therapeutic assays that examine conditions associated with dysregulated cell growth such as cancer, in particular the cancers listed in Table I (see, both its specific pattern of tissue expression as well as its overexpression in certain cancers as described for example in the Example entitled "Expression analysis of PSCA in normal tissues, and patient specimens").
[0288] PSCA can be analogized to a prostate associated antigen PSA, the archetypal marker that has been used by medical practitioners for years to identify and monitor the presence of prostate cancer (see, Merrill et al., J. Urol.
163(2): 503-5120 (2000); Polascik et al., J. Urol. Aug; 162(2):293-306 (1999) and Fortier et aL, J. Nat Cancer Inst. 91(19): 1635-1640(1999)). A variety of other diagnostic markers are also used in similar contexts including p53 and K-ras (see, Tulchinsky et Int J Mol Med 1999 Jul 4(1):99-102 and Minimoto et Cancer Detect Prev 2000;24(1):1-12).
Therefore, this disclosure of PSCA polynucleotides and polypeptides (as well as PSCA polynucleotide probes and anti- PSCA antibodies used to identify the presence of these molecules) and their properties allows skilled artisans to utilize these molecules in methods that are analogous to those used, for example, in a variety of diagnostic assays directed to examining conditions associated with cancer.
[0289] Typical embodiments of diagnostic methods which utilize the PSCA polynucleotides, polypeptides, reactive T cells and antibodies are analogous to those methods from well-established diagnostic assays, which employ, PSA polynucleotides, polypeptides, reactive T cells and antibodies. For example, just as PSA polynucleotides are used as probes (for example in Northern analysis, see, Sharief et al., Biochem. Mol. Biol. Int. 33(3):567-74(1994)) and primers (for example in PCR analysis, see, Okegawa et aL, J. Urol. 163(4): 1189-1190 (2000)) to observe the presence and/or the level of PSA mRNAs in methods of monitoring PSA overexpression or the metastasis of prostate cancers, the PSCA polynucleotides described herein can be utilized in the same way to detect PSCA overexpression or the metastasis of prostate and other cancers expressing this gene. Alternatively, just as PSA polypeptides are used to generate antibodies specific for PSA which can then be used to observe the presence and/or the level of PSA proteins in methods to monitor PSA protein overexpression (see, Stephan et al., Urology 55(4):560-3 (2000)) or the metastasis of prostate cells (see, Alanen et Pathol. Res. Pract. 192(3):233-7 (1996)), the PSCA polypeptides described herein can be utilized to generate antibodies for use in detecting PSCA overexpression or the metastasis of prostate cells and cells of other cancers expressing this gene.
[0290] Specifically, because metastases involves the movement of cancer cells from an organ of origin (such as the lung or prostate gland etc.) to a different area of the body (such as a lymph node), assays which examine a biological sample for the presence of cells expressing PSCA polynucleotides and/or polypeptides can be used to provide evidence of metastasis. For example, when a biological sample from tissue that does not normally contain PSCA-expressing cells (lymph node) is found to contain PSCA-expressing cells such as the PSCA expression seen in LAPC4 and LAPC9, xenografts isolated from lymph node and bone metastasis, respectively, this finding is indicative of metastasis.
[0291] Alternatively PSCA polynucleotides and/or polypeptides can be used to provide evidence of cancer, for example, when cells in a biological sample that do not normally express PSCA or express PSCA at a different level are found to express PSCA or have an increased expression of PSCA (see, the PSCA expression in the cancers listed in Table I and in patient samples etc. shown in the accompanying Figures). In such assays, artisans may further wish to generate supplementary evidence of metastasis by testing the biological sample for the presence of a second tissue WO 2005/014780 PCT/US2004/017231 restricted marker (in addition to PSCA) such as PSA, PSCA etc. (see, Alanen e al., Pathol. Res. Pract. 192(3): 233- 237 (1996)).
[0292] The use of immunohistochemistry to identify the presence of a PSCA polypeptide within a tissue section can indicate an altered state of certain cells within that tissue. It is well understood in the art that the ability of an antibody to localize to a polypeptide that is expressed in cancer cells is a way of diagnosing presence of disease, disease stage, progression and/or tumor aggressiveness. Such an antibody can also detect an altered distribution of the polypeptide within the cancer cells, as compared to corresponding non-malignant tissue.
[0293] The PSCA polypeptide and immunogenic compositions are also useful in view of the phenomena of altered subcellular protein localization in disease states. Alteration of cells from normal to diseased state causes changes in cellular morphology and is often associated with changes in subcellular protein localization/distribution. For example, cell membrane proteins that are expressed in a polarized manner in normal cells can be altered in disease, resulting in distribution of the protein in a non-polar manner over the whole cell surface.
[0294] The phenomenon of altered subcellular protein localization in a disease state has been demonstrated with MUC1 and Her2 protein expression by use of immunohistochemical means. Normal epithelial cells have a typical apical distribution of MUC1, in addition to some supranuclear localization of the glycoprotein, whereas malignant lesions often demonstrate an apolar staining pattern (Diaz et al, The Breast Journal, 7; 40-45 (2001); Zhang et al, Clinical Cancer Research, 4; 2669-2676 (1998): Cao, et al, The Journal of Histochemistry and Cytochemistry, 45:1547-1557 (1997)). In addition, normal breast epithelium is either negative for Her2 protein or exhibits only a basolateral distribution whereas malignant cells can express the protein over the whole cell surface (De Potter, et al, International Journal of Cancer, 44; 969-974 (1989): McCormick, et al, 117; 935-943 (2002)). Alternatively, distribution of the protein may be altered from a surface only localization to include diffuse cytoplasmic expression in the diseased state. Such an example can be seen with MUC1 (Diaz, et al, The Breast Journal, 7: 40-45 (2001)).
[0295] Alteration in the localization/distribution of a protein in the cell, as detected by immunohistochemical methods, can also provide valuable information concerning the favorability of certain treatment modalities. This last point is illustrated by a situation where a protein may be intracellular in normal tissue, but cell surface in malignant cells; the cell surface location makes the cells favorably amenable to antibody-based diagnostic and treatment regimens. When such an alteration of protein localization occurs for PSCA, the PSCA protein and immune responses related thereto are very useful.
Accordingly, the ability to determine whether alteration of subcellular protein localization occurred for 24P4C12 make the PSCA protein and immune responses related thereto very useful. Use of the PSCA compositions allows those skilled in the art to make important diagnostic and therapeutic decisions.
[0296] Immunohistochemical reagents specific to PSCA are also useful to detect metastases of tumors expressing PSCA when the polypeptide appears in tissues where PSCA is not normally produced.
[0297] Thus, PSCA polypeptides and antibodies resulting from immune responses thereto are useful in a variety of important contexts such as diagnostic, prognostic, preventative and/or therapeutic purposes known to those skilled in the art.
[0298] Just as PSA polynucleotide fragments and polynucleotide variants are employed by skilled artisans for use in methods of monitoring PSA, PSCA polynucleotide fragments and polynucleotide variants are used in an analogous manner. In particular, typical PSA polynucleotides used in methods of monitoring PSA are probes or primers which consist of fragments of the PSA cDNA sequence. Illustrating this, primers used to PCR amplify a PSA polynucleotide must include less than the whole PSA sequence to function in the polymerase chain reaction. In the context of such PCR reactions, 68 WO 2005/014780 PCT/US2004/017231 skilled artisans generally create a variety of different polynucleotide fragments that can be used as primers in order to amplify different portions of a polynucleotide of interest or to optimize amplification reactions (see, Caetano-Anolles, G.
Biotechniques 25(3): 472-476, 478-480 (1998); Robertson et Methods Mol. Biol. 98:121-154 (1998)). An additional illustration of the use of such fragments is provided in the Example entitled "Expression analysis of PSCA in normal tissues, and patient specimens," where a PSCA polynucleotide fragment is used as a probe to show the expression of PSCA RNAs in cancer cells. In addition, variant polynucleotide sequences are typically used as primers and probes for the corresponding mRNAs in PCR and Northern analyses (see, Sawai et al, Fetal Diagn. Ther. 1996 Nov-Dec 11(6):407- 13 and Current Protocols In Molecular Biology, Volume 2, Unit 2, Frederick M. Ausubel et a. eds., 1995)). Polynucleotide fragments and variants are useful in this context where they are capable of binding to a target polynucleotide sequence a PSCA polynucleotide shown in Figure 2 or variant thereof) under conditions of high stringency.
[0299] Furthermore, PSA polypeptides which contain an epitope that can be recognized by an antibody or T cell that specifically binds to that epitope are used in methods of monitoring PSA. PSCA polypeptide fragments and polypeptide analogs or variants can also be used in an analogous manner. This practice of using polypeptide fragments or polypeptide variants to generate antibodies (such as anti-PSA antibodies or T cells) is typical in the art with a wide variety of systems such as fusion proteins being used by practitioners (see, Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubel et al. eds., 1995). In this context, each epitope(s) functions to provide the architecture with which an antibody or T cell is reactive. Typically, skilled artisans create a variety of different polypeptide fragments that can be used in order to generate immune responses specific for different portions of a polypeptide of interest (see, U.S. Patent No.
5,840,501 and U.S. Patent No. 5,939,533). For example it may be preferable to utilize a polypeptide comprising one of the PSCA biological motifs discussed herein or a motif-bearing subsequence which is readily identified by one of skill in the art based on motifs available in the art. Polypeptide fragments, variants or analogs are typically useful in this context as long as they comprise an epitope capable of generating an antibody or T cell specific for a target polypeptide sequence a PSCA polypeptide shown in Figure 3).
[0300] As shown herein, the PSCA polynucleotides and polypeptides (as well as the PSCA polynucleotide probes and anti-PSCA antibodies or T cells used to identify the presence of these molecules) exhibit specific properties that make them useful in diagnosing cancers such as those listed in Table I. Diagnostic assays that measure the presence of PSCA gene products, in order to evaluate the presence or onset of a disease condition described herein, such as prostate cancer, are used to identify patients for preventive measures or further monitoring, as has been done so successfully with PSA.
Moreover, these materials satisfy a need in the art for molecules having similar or complementary characteristics to PSA in situations where, for example, a definite diagnosis of metastasis of prostatic origin cannot be made on the basis of a test for PSA alone (see, Alanen et al., Pathol. Res. Pract. 192(3): 233-237 (1996)), and consequently, materials such as PSCA polynucleotides and polypeptides (as well as the PSCA polynucleotide probes and anti-PSCA antibodies used to identify the presence of these molecules) need to be employed to confirm a metastases of prostatic origin.
[0301] Finally, in addition to their use in diagnostic assays, the PSCA polynucleotides disclosed herein have a number of other utilities such as their use in the identification of oncogenetic associated chromosomal abnormalities in the chromosomal region to which the PSCA gene maps (see the Example entitled "Chromosomal Mapping of PSCA" below).
Moreover, in addition to their use in diagnostic assays, the PSCA-related proteins and polynucleotides disclosed herein have other utilities such as their use in the forensic analysis of tissues of unknown origin (see, Takahama K Forensic Sci Int 1996 Jun 28;80(1-2): 63-9).
WO 2005/014780 PCT/US2004/017231 [0302] Additionally, PSCA-related proteins or polynucleotides of the invention can be used to treat a pathologic condition characterized by the over-expression of PSCA. For example, the amino acid or nucleic acid sequence of Figure 2 or Figure 3, or fragments of either, can be used to generate an immune response to a PSCA antigen. Antibodies or other molecules that react with PSCA can be used to modulate the function of this molecule, and thereby provide a therapeutic benefit.
XII.) Inhibition of PSCA Protein Function [0303] The invention includes various methods and compositions for inhibiting the binding of PSCA to its binding partner or its association with other protein(s) as well as methods for inhibiting PSCA function.
XII.A.) Inhibition of PSCA With Intracellular Antibodies [0304] In one approach, a recombinant vector that encodes single chain antibodies that specifically bind to PSCA are introduced into PSCA expressing cells via gene transfer technologies. Accordingly, the encoded single chain anti- PSCA antibody is expressed intracellularly, binds to PSCA protein, and thereby inhibits its function. Methods for engineering such intracellular single chain antibodies are well known. Such intracellular antibodies, also known as "intrabodies", are specifically targeted to a particular compartment within the cell, providing control over where the inhibitory activity of the treatment is focused. This technology has been successfully applied in the art (for review, see Richardson and Marasco, 1995, TIBTECH vol. 13). Intrabodies have been shown to virtually eliminate the expression of otherwise abundant cell surface receptors (see, Richardson et 1995, Proc. Natl. Acad. Sci. USA 92: 3137-3141; Beerli et a., 1994, J. Biol. Chem. 289: 23931-23936; Deshane et al., 1994, Gene Ther. 1:332-337).
[0305] Single chain antibodies comprise the variable domains of the heavy and light chain joined by a flexible linker polypeptide, and are expressed as a single polypeptide. Optionally, single chain antibodies are expressed as a single chain variable region fragment joined to the light chain constant region. Well-known intracellular trafficking signals are engineered into recombinant polynucleotide vectors encoding such single chain antibodies in order to target precisely the intrabody to the desired intracellular compartment. For example, intrabodies targeted to the endoplasmic reticulum (ER) are engineered to incorporate a leader peptide and, optionally, a C-terminal ER retention signal, such as the KDEL amino acid motif. Intrabodies intended to exert activity in the nucleus are engineered to include a nuclear localization signal. Llpid moieties are joined to intrabodies in order to tether the intrabody to the cytosolic side of the plasma membrane. Intrabodies can also be targeted to exert function in the cytosol. For example, cytosolic intrabodies are used to sequester factors within the cytosol, thereby preventing them from being transported to their natural cellular destination.
[0306] In one embodiment, intrabodies are used to capture PSCA in the nucleus, thereby preventing its activity within the nucleus. Nuclear targeting signals are engineered into such PSCA intrabodies in order to achieve the desired targeting. Such PSCA intrabodies are designed to bind specifically to a particular PSCA domain. In another embodiment, cytosolic intrabodies that specifically bind to a PSCA protein are used to prevent PSCA from gaining access to the nucleus, thereby preventing it from exerting any biological activity within the nucleus preventing PSCA from forming transcription complexes with other factors).
[0307] In order to specifically direct the expression of such intrabodies to particular cells, the transcription of the intrabody is placed under the regulatory control of an appropriate tumor-specific promoter andlor enhancer. In order to target intrabody expression specifically to prostate, for example, the PSA promoter and/or promoter/enhancer can be utilized (See, for example, U.S. Patent No. 5,919,652 issued 6 July 1999).
WO 2005/014780 PCT/US2004/017231 XII.B.) Inhibition of PSCA with Recombinant Proteins [0308] In another approach, recombinant molecules bind to PSCA and thereby inhibit PSCA function. For example, these recombinant molecules prevent or inhibit PSCA from accessing/binding to its binding partner(s) or associating with other protein(s). Such recombinant molecules can, for example, contain the reactive part(s) of a PSCA specific antibody molecule. In a particular embodiment, the PSCA binding domain of a PSCA binding partner is engineered into a dimeric fusion protein, whereby the fusion protein comprises two PSCA ligand binding domains linked to the Fc portion of a human IgG, such as human IgG1. Such igG portion can contain, for example, the CH 2 and CH3 domains and the hinge region, but not the CH1 domain. Such dimeric fusion proteins are administered in soluble form to patients suffering from a cancer associated with the expression of PSCA, whereby the dimeric fusion protein specifically binds to PSCA and blocks PSCA interaction with a binding partner. Such dimeric fusion proteins are further combined into multimeric proteins using known antibody linking technologies.
XII.C.) Inhibition of PSCA Transcription or Translation [0309] The present invention also comprises various methods and compositions for inhibiting the transcription of the PSCA gene. Similarly, the invention also provides methods and compositions for inhibiting the translation of PSCA mRNA into protein.
[0310] In one approach, a method of inhibiting the transcription of the PSCA gene comprises contacting the PSCA gene with a PSCA antisense polynucleotide. In another approach, a method of inhibiting PSCA mRNA translation comprises contacting a PSCA mRNA with an antisense polynucleotide. In another approach, a PSCA specific ribozyme is used to cleave a PSCA message, thereby inhibiting translation. Such antisense and ribozyme based methods can also be directed to the regulatory regions of the PSCA gene, such as PSCA promoter and/or enhancer elements. Similarly, proteins capable of inhibiting a PSCA gene transcription factor are used to inhibit PSCA mRNA transcription. The various polynucleotides and compositions useful in the aforementioned methods have been described above. The use of antisense and ribozyme molecules to inhibit transcription and translation is well known in the art.
[0311] Other factors that inhibit the transcription of PSCA by interfering with PSCA transcriptional activation are also useful to treat cancers expressing PSCA. Similarly, factors that interfere with PSCA processing are useful to treat cancers that express PSCA. Cancer treatment methods utilizing such factors are also within the scope of the invention.
XII.D.) General Considerations for Therapeutic Strategies [0312] Gene transfer and gene therapy technologies can be used to deliver therapeutic polynucleotide molecules to tumor cells synthesizing PSCA antisense, ribozyme, polynucleotides encoding intrabodies and other PSCA inhibitory molecules). A number of gene therapy approaches are known in the art. Recombinant vectors encoding PSCA antisense polynucleotides, ribozymes, factors capable of interfering with PSCA transcription, and so forth, can be delivered to target tumor cells using such gene therapy approaches.
[0313] The above therapeutic approaches can be combined with any one of a wide variety of surgical, chemotherapy or radiation therapy regimens. The therapeutic approaches of the invention can enable the use of reduced dosages of chemotherapy (or other therapies) and/or less frequent administration, an advantage for all patients and particularly for those that do not tolerate the toxicity of the chemotherapeutic agent well.
[0314] The anti-tumor activity of a particular composition antisense, ribozyme, intrabody), or a combination of such compositions, can be evaluated using various in vitro and in vivo assay systems. In vitro assays that evaluate WO 2005/014780 PCT/US2004/017231 therapeutic activity include cell growth assays, soft agar assays and other assays indicative of tumor promoting activity, binding assays capable of determining the extent to which a therapeutic composition will inhibit the binding of PSCA to a binding partner, etc.
[0315] In vivo, the effect of a PSCA therapeutic composition can be evaluated in a suitable animal model. For example, xenogenic prostate cancer models can be used, wherein human prostate cancer explants or passaged xenograft tissues are introduced into immune compromised animals, such as nude or SCID mice (Klein et al., 1997, Nature Medicine 3: 402-408). For example, PCT Patent Application W098/16628 and U.S. Patent 6,107,540 describe various xenograft models of human prostate cancer capable of recapitulating the development of primary tumors, micrometastasis, and the formation ofosteoblastic metastases characteristic of late stage disease. Efficacy can be predicted using assays that measure inhibition of tumor formation, tumor regression or metastasis, and the like.
[0316] In vivo assays that evaluate the promotion of apoptosis are useful in evaluating therapeutic compositions. In one embodiment, xenografts from tumor bearing mice treated with the therapeutic composition can be examined for the presence of apoptotic foci and compared to untreated control xenograft-bearing mice. The extent to which apoptotic foci are found in the tumors of the treated mice provides an indication of the therapeutic efficacy of the composition.
[0317] The therapeutic compositions used in the practice of the foregoing methods can be formulated into pharmaceutical compositions comprising a carrier suitable for the desired delivery method. Suitable carriers include any material that when combined with the therapeutic composition retains the anti-tumor function of the therapeutic composition and is generally non-reactive with the patient's immune system. Examples include, but are not limited to, any of a number of standard pharmaceutical carriers such as sterile phosphate buffered saline solutions, bacteriostatic water, and the like (see, generally, Remington's Pharmaceutical Sciences 16 t Edition, A. Osal., Ed., 1980).
[0318] Therapeutic formulations can be solubilized and administered via any route capable of delivering the therapeutic composition to the tumor site. Potentially effective routes of administration include, but are not limited to, intravenous, parenteral, intraperitoneal, intramuscular, intratumor, intradermal, intraorgan, orthotopic, and the like. A preferred formulation for intravenous injection comprises the therapeutic composition in a solution of preserved bacteriostatic water, sterile unpreserved water, and/or diluted in polyvinylchloride or polyethylene bags containing 0.9% sterile Sodium Chloride for Injection, USP. Therapeutic protein preparations can be lyophilized and stored as sterile powders, preferably under vacuum, and then reconstituted in bacteriostatic water (containing for example, benzyl alcohol preservative) or in sterile water prior to injection.
[0319] Dosages and administration protocols for the treatment of cancers using the foregoing methods will vary with the method and the target cancer, and will generally depend on a number of other factors appreciated in the art.
XIII.) Identification. Characterization and Use of Modulators of PSCA Methods to Identify and Use Modulators [0320] In one embodiment, screening is performed to identify modulators that induce or suppress a particular expression profile, suppress or induce specific pathways, preferably generating the associated phenotype thereby. In another embodiment, having identified differentially expressed genes important in a particular state; screens are performed to identify modulators that alter expression of individual genes, either increase or decrease. In another embodiment, screening is performed to identify modulators that alter a biological function of the expression product of a differentially WO 2005/014780 PCT/LS2004/017231 expressed gene. Again, having identified the importance of a gene in a particular state, screens are performed to identify agents that bind and/or modulate the biological activity of the gene product.
[0321] In addition, screens are done for genes that are induced in response to a candidate agent. After identifying a modulator (one that suppresses a cancer expression pattern leading to a normal expression pattern, or a modulator of a cancer gene that leads to expression of the gene as in normal tissue) a screen is performed to identify genes that are specifically modulated in response to the agent. Comparing expression profiles between normal tissue and agent-treated cancer tissue reveals genes that are not expressed in normal tissue or cancer tissue, but are expressed in agent treated tissue, and vice versa. These agent-specific sequences are identified and used by methods described herein for cancer genes or proteins. In particular these sequences and the proteins they encode are used in marking or identifying agenttreated cells. In addition, antibodies are raised against the agent-induced proteins and used to target novel therapeutics to the treated cancer tissue sample.
Modulator-related Identification and Screening Assays: Gene Expression-related Assays [0322] Proteins, nucleic acids, and antibodies of the invention are used in screening assays. The cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing these sequences are used in screening assays, such as evaluating the effect of drug candidates on a "gene expression profile," expression profile of polypeptides or alteration of biological function. In one embodiment, the expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring for expression profile genes after treatment with a candidate agent Davis, GF, et al, J Biol Screen 7:69 (2002); Zlokarnik, et al., Science 279:84-8 (1998); Heid, Genome Res 6:986- 94,1996).
[0323] The cancer proteins, antibodies, nucleic acids, modified proteins and cells containing the native or modified cancer proteins or genes are used in screening assays. That is, the present invention comprises methods for screening for compositions which modulate the cancer phenotype or a physiological function of a cancer protein of the invention. This is done on a gene itself or by evaluating the effect of drug candidates on a "gene expression profile" or biological function. In one embodiment, expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring after treatment with a candidate agent, see Zlokamik, supra.
[0324] A variety of assays are executed directed to the genes and proteins of the invention. Assays are run on an individual nucleic acid or protein level. That is, having identified a particular gene as up regulated in cancer, test compounds are screened for the ability to modulate gene expression or for binding to the cancer protein of the invention.
"Modulation" in this context includes an increase or a decrease in gene expression. The preferred amount of modulation will depend on the original change of the gene expression in normal versus tissue undergoing cancer, with changes of at least 10%, preferably 50%, more preferably 100-300%, and in some embodiments 300-1000% or greater. Thus, if a gene exhibits a 4-fold increase in cancer tissue compared to normal tissue, a decrease of about four-fold is often desired; similarly, a 10-fold decrease in cancer tissue compared to normal tissue a target value of a 10-fold increase in expression by the test compound is often desired. Modulators that exacerbate the type of gene expression seen in cancer are also useful, as an upregulated target in further analyses.
WO 2005/014780 PCT/US2004/017231 [0325] The amount of gene expression is monitored using nucleic acid probes and the quantification of gene expression levels, or, alternatively, a gene product itself is monitored, through the use of antibodies to the cancer protein and standard immunoassays. Proteomics and separation techniques also allow for quantification of expression.
Expression Monitoring to Identify Compounds that Modify Gene Expression [0326] In one embodiment, gene expression monitoring, an expression profile, is monitored simultaneously for a number of entities. Such profiles will typically involve one or more of the genes of Figure 2. In this embodiment, e.g., cancer nucleic acid probes are attached to biochips to detect and quantify cancer sequences in a particular cell.
Alternatively, PCR can be used. Thus, a series, wells of a microtiter plate, can be used with dispensed primers in desired wells. A PCR reaction can then be performed and analyzed for each well.
[0327] Expression monitoring is performed to identify compounds that modify the expression of one or more cancerassociated sequences, a polynucleotide sequence set out in Figure 2. Generally, a test modulator is added to the cells prior to analysis. Moreover, screens are also provided to identify agents that modulate cancer, modulate cancer proteins of the invention, bind to a cancer protein of the invention, or interfere with the binding of a cancer protein of the invention and an antibody or other binding partner.
[0328] In one embodiment, high throughput screening methods involve providing a library containing a large number of potential therapeutic compounds (candidate compounds). Such "combinatorial chemical libraries" are then screened in one or more assays to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional "lead compounds," as compounds for screening, or as therapeutics.
[0329] In certain embodiments, combinatorial libraries of potential modulators are screened for an ability to bind to a cancer polypeptide or to modulate activity. Conventionally, new chemical entities with useful properties are generated by identifying a chemical.compound (called a "lead compound") with some desirable property or activity, inhibiting activity, creating variants of the lead compound, and evaluating the property and activity of those variant compounds. Often, high throughput screening (HTS) methods are employed for such an analysis.
[0330] As noted above, gene expression monitoring is conveniently used to test candidate modulators protein, nucleic acid or small molecule). After the candidate agent has been added and the cells allowed to incubate for a period, the sample containing a target sequence to be analyzed is, added to a biochip.
[0331] If required, the target sequence is prepared using known techniques. For example, a sample is treated to lyse the cells, using known lysis buffers, electroporation, etc., with purification and/or amplification such as PCR performed as appropriate. For example, an in vitro transcription with labels covalently attached to the nucleotides is performed.
Generally, the nucleic acids are labeled with biotin-FITC or PE, or with cy3 or [0332] The target sequence can be labeled with, a fluorescent, a chemiluminescent, a chemical, or a radioactive signal, to provide a means of detecting the target sequence's specific binding to a probe. The label also can be an enzyme, such as alkaline phosphatase or horseradish peroxidase, which when provided with an appropriate substrate produces a product that is detected. Alternatively, the label is a labeled compound or small molecule, such as an enzyme inhibitor, that binds but is not catalyzed or altered by the enzyme. The label also can be a moiety or compound, such as, an epitope tag or biotin which specifically binds to streptavidin. For the example of biotin, the streptavidin is labeled as described above, thereby, providing a detectable signal for the bound target sequence. Unbound labeled streptavidin is typically removed prior to analysis.
WO 2005/014780 PCT/US2004/017231 [0333] As will be appreciated by those in the art, these assays can be direct hybridization assays or can comprise "sandwich assays", which include the use of multiple probes, as is generally outlined in U.S. Patent Nos. 5, 681,702; 5,597,909; 5,545,730; 5,594,117; 5,591,584; 5,571,670; 5,580,731; 5,571,670; 5,591,584; 5,624,802; 5,635,352; 5,594,118; 5,359,100; 5,124, 246; and 5,681,697. In this embodiment, in general, the target nucleic acid is prepared as outlined above, and then added to the biochip comprising a plurality of nucleic acid probes, under conditions that allow the formation of a hybridization complex.
[0334] A variety of hybridization conditions are used in the present invention, including high, moderate and low stringency conditions as outlined above. The assays are generally run under stringency conditions which allow formation of the label probe hybridization complex only in the presence of target. Stringency can be controlled by altering a step parameter that is a thermodynamic variable, including, but not limited to, temperature, formamide concentration, salt concentration, chaotropic salt concentration pH, organic solvent concentration, etc. These parameters may also be used to control non-specific binding, as is generally outlined in U.S. Patent No. 5,681,697. Thus, it can be desirable to perform certain steps at higher stringency conditions to reduce non-specific binding.
[0335] The reactions outlined herein can be accomplished in a variety of ways. Components of the reaction can be added simultaneously, or sequentially, in different orders, with preferred embodiments outlined below. In addition, the reaction may include a variety of other reagents. These include salts, buffers, neutral proteins, e.g. albumin, detergents, etc,. which can be used to facilitate optimal hybridization and detection, and/or reduce nonspecific or background interactions. Reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may also be used as appropriate, depending on the sample preparation methods and purity of the target. The assay data are analyzed to determine the expression levels of individual genes, and changes in expression levels as between states, forming a gene expression profile.
Biological Activity-related Assays [0336] The invention provides methods identify or screen for a compound that modulates the activity of a cancerrelated gene or protein of the invention. The methods comprise adding a test compound, as defined above, to a cell comprising a cancer protein of the invention. The cells contain a recombinant nucleic acid that encodes a cancer protein of the invention. In another embodiment, a library of candidate agents is tested on a plurality of cells.
[0337] In one aspect, the assays are evaluated in the presence or absence or previous or subsequent exposure of physiological signals, e.g. hormones, antibodies, peptides, antigens, cytokines, growth factors, action potentials, pharmacological agents including chemotherapeutics, radiation, carcinogenics, or other cells cell-cell contacts). In another example, the determinations are made at different stages of the cell cycle process. In this way, compounds that modulate genes or proteins of the invention are identified. Compounds with pharmacological activity are able to enhance or interfere with the activity of the cancer protein of the invention. Once identified, similar structures are evaluated to identify critical structural features of the compound.
[0338] In one embodiment, a method of modulating inhibiting) cancer cell division is provided; the method comprises administration of a cancer modulator. In another embodiment, a method of modulating inhibiting) cancer is provided; the method comprises administration of a cancer modulator. In a further embodiment, methods of treating cells or individuals with cancer are provided; the method comprises administration of a cancer modulator.
[0339] In one embodiment, a method for modulating the status of a cell that expresses a gene of the invention is provided. As used herein status comprises such art-accepted parameters such as growth, proliferation, survival, function, WO 2005/014780 PCT/LS2004/017231 apoptosis, senescence, location, enzymatic activity, signal transduction, etc. of a cell. In one embodiment, a cancer inhibitor is an antibody as discussed above. In another embodiment, the cancer inhibitor is an antisense molecule. A variety of cell growth, proliferation, and metastasis assays are known to those of skill in the art, as described herein.
High Throughput Screening to Identify Modulators [0340] The assays to identify suitable modulators are amenable to high throughput screening. Preferred assays thus detect enhancement or inhibition of cancer gene transcription, inhibition or enhancement of polypeptide expression, and inhibition or enhancement of polypeptide activity.
[0341] In one embodiment, modulators evaluated in high throughput screening methods are proteins, often naturally occurring proteins or fragments of naturally occurring proteins. Thus, cellular extracts containing proteins, or random or directed digests of proteinaceous cellular extracts, are used. In this way, libraries of proteins are made for screening in the methods of the invention. Particularly preferred in this embodiment are libraries of bacterial, fungal, viral, and mammalian proteins, with the latter being preferred, and human proteins being especially preferred. Particularly useful test compound will be directed to the class of proteins to which the target belongs, substrates for enzymes, or ligands and receptors.
Use of Soft Aqar Growth and Colony Formation to Identify and Characterize Modulators [0342] Normal cells require a solid substrate to attach and grow. When cells are transformed, they lose this phenotype and grow detached from the substrate. For example, transformed cells can grow in stirred suspension culture or suspended in semi-solid media, such as semi-solid or soft agar. The transformed cells, when transfected with tumor suppressor genes, can regenerate normal phenotype and once again require a solid substrate to attach to and grow. Soft agar growth or colony formation in assays are used to identify modulators of cancer sequences, which when expressed in host cells, inhibit abnormal cellular proliferation and transformation. A modulator reduces or eliminates the host cells' ability to grow suspended in solid or semisolid media, such as agar.
[0343] Techniques for soft agar growth or colony formation in suspension assays are described in Freshney, Culture of Animal Cells a Manual of Basic Technique (3rd ed., 1994). See also, the methods section of Garkavtsev et al. (1996), supra.
Evaluation of Contact Inhibition and Growth Density Limitation to Identify and Characterize Modulators [0344] Normal cells typically grow in a flat and organized pattern in cell culture until they touch other cells. When the cells touch one another, they are contact inhibited and stop growing. Transformed cells, however, are not contact inhibited and continue to grow to high densities in disorganized foci. Thus, transformed cells grow to a higher saturation density than corresponding normal cells. This is detected morphologically by the formation of a disoriented monolayer of cells or cells in foci. Alternatively, labeling index with 3 H)-thymidine at saturation density is used to measure density limitation of growth, similarly an MTT or Alamar blue assay will reveal proliferation capacity of cells and the the ability of modulators to affect same. See Freshney (1994), supra. Transformed cells, when transfected with tumor suppressor genes, can regenerate a normal phenotype and become contact inhibited and would grow to a lower density.
[0345] In this assay, labeling index with 3 H)-thymidine at saturation density is a preferred method of measuring density limitation of growth. Transformed host cells are transfected with a cancer-associated sequence and are grown for 76 WO 2005/014780 PCT/US2004/017231 24 hours at saturation density in non-limiting medium conditions. The percentage of cells labeling with 3 H)-thymidine is determined by incorporated cpm.
[0346] Contact independent growth is used to identify modulators of cancer sequences, which had led to abnormal cellular proliferation and transformation. A modulator reduces or eliminates contact independent growth, and returns the cells to a normal phenotype.
Evaluation of Growth Factor or Serum Dependence to Identify and Characterize Modulators [0347] Transformed cells have lower serum dependence than their normal counterparts (see, Temin, J. Natl.
Cancer Inst. 37:167-175 (1966); Eagle et al., J. Exp. Med 131:836-879 (1970)); Freshney, supra. This is in part due to release of various growth factors by the transformed cells. The degree of growth factor or serum dependence of transformed host cells can be compared with that of control. For example, growth factor or serum dependence of a cell is monitored in methods to identify and characterize compounds that modulate cancer-associated sequences of the invention.
Use of Tumor-specific Marker Levels to Identify and Characterize Modulators [0348] Tumor cells release an increased amount of certain factors (hereinafter "tumor specific markers") than their normal counterparts. For example, plasminogen activator (PA) is released from human glioma at a higher level than from normal brain cells (see, Gullino, Angiogenesis, Tumor Vascularization, and Potential Interference with Tumor Growth, in Biological Responses in Cancer, pp. 178-184 (Mihich 1985)). Similarly, Tumor Angiogenesis Factor (TAF) is released at a higher level in tumor cells than their normal counterparts. See, Folkman, Angiogenesis and Cancer, Sem Cancer Biol. (1992)), while bFGF is released from endothelial tumors (Ensoli, B et al).
[0349] Various techniques which measure the release of these factors are described in Freshney (1994), supra.
Also, see, Unkless et al., J. Biol. Chem. 249:4295-4305 (1974); Strickland Beers, J. Biol. Chem. 251:5694-5702 (1976); Whur et al., Br. J. Cancer 42:305 312 (1980); Gullino, Angiogenesis, Tumor Vascularization, and Potential Interference with Tumor Growth, in Biological Responses in Cancer, pp. 178-184 (Mihich 1985); Freshney, Anticancer Res. 5:111-130 (1985). For example, tumor specific marker levels are monitored in methods to identify and characterize compounds that modulate cancer-associated sequences of the invention.
Invasiveness into Matrigel to Identify and Characterize Modulators [0350] The degree of invasiveness into Matrigel or an extracellular matrix constituent can be used as an assay to identify and characterize compounds that modulate cancer associated sequences. Tumor cells exhibit a positive correlation between malignancy and invasiveness of cells into Matrigel or some other extracellular matrix constituent. In this assay, tumorigenic cells are typically used as host cells. Expression of a tumor suppressor gene in these host cells would decrease invasiveness of the host cells. Techniques described in Cancer Res. 1999; 59:6010; Freshney (1994), supra, can be used. Briefly, the level of invasion of host cells is measured by using filters coated with Matrigel or some other extracellular matrix constituent. Penetration into the gel, or through to the distal side of the filter, is rated as invasiveness, and rated histologically by number of cells and distance moved, or by prelabeling the cells with 1251 and counting the radioactivity on the distal side of the filter or bottom of the dish. See, Freshney (1984), supra.
WO 2005/014780 PCT/US2004/017231 Evaluation of Tumor Growth In Vivo to Identify and Characterize Modulators [0351] Effects of cancer-associated sequences on cell growth are tested in transgenic or immune-suppressed organisms. Transgenic organisms are prepared in a variety of art-accepted ways. For example, knock-out transgenic organisms, mammals such as mice, are made, in which a cancer gene is disrupted or in which a cancer gene is inserted. Knock-out transgenic mice are made by insertion of a marker gene or other heterologous gene into the endogenous cancer gene site in the mouse genome via homologous recombination. Such mice can also be made by substituting the endogenous cancer gene with a mutated version of the cancer gene, or by mutating the endogenous cancer gene, by exposure to carcinogens.
[0352] To prepare transgenic chimeric animals, mice, a DNA construct is introduced into the nuclei of embryonic stem cells. Cells containing the newly engineered genetic lesion are injected into a host mouse embryo, which is re-implanted into a recipient female. Some of these embryos develop into chimeric mice that possess germ cells some of which are derived from the mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion (see, Capecchi et al., Science 244:1288 (1989)). Chimeric mice can be derived according to US Patent 6,365,797, issued 2 April 2002; US Patent 6,107,540 issued 22 August 2000; Hogan et al., Manipulating the Mouse Embryo: A laboratory Manual, Cold Spring Harbor Laboratory (1988) and Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Robertson, ed., IRL Press, Washington, (1987).
[0353] Alternatively, various immune-suppressed or immune-deficient host animals can be used. For example, a genetically athymic "nude" mouse (see, Giovanella et al., J. Natl. Cancer Inst. 52:921 (1974)), a SCID mouse, a thymectornized mouse, or an irradiated mouse (see, Bradley et al., Br. J. Cancer 38:263 (1978); Selby et al., Br. J.
Cancer 41:52 (1980)) can be used as a host. Transplantable tumor cells (typically about 106 cells) injected into isogenic hosts produce invasive tumors in a high proportion of cases, while normal cells of similar origin will not. In hosts which developed invasive tumors, cells expressing cancer-associated sequences are injected subcutaneously or orthotopically.
Mice are then separated into groups, including control groups and treated experimental groups) e.g. treated with a modulator). After a suitable length of time, preferably 4-8 weeks, tumor growth is measured by volume or by its two largest dimensions, or weight) and compared to the control. Tumors that have statistically significant reduction (using, e.g., Student's T test) are said to have inhibited growth.
In Vitro Assays to Identify and Characterize Modulators [0354] Assays to identify compounds with modulating activity can be performed in vitro. For example, a cancer polypeptide is first contacted with a potential modulator and incubated for a suitable amount of time, from 0.5 to 48 hours. In one embodiment, the cancer polypeptide levels are determined in vitro by measuring the level of protein or mRNA. The level of protein is measured using immunoassays such as Western blotting, ELISA and the like with an antibody that selectively binds to the cancer polypeptide or a fragment thereof. For measurement of mRNA, amplification, using PCR, LCR, or hybridization assays, e. Northern hybridization, RNAse protection, dot blotting, are preferred.
The level of protein or mRNA is detected using directly or indirectly labeled detection agents, fluorescently or radioactively labeled nucleic acids, radioactively or enzymatically labeled antibodies, and the like, as described herein.
[0355] Alternatively, a reporter gene system can be devised using a cancer protein promoter operably linked to a reporter gene such as luciferase, green fluorescent protein, CAT, or P-gal. The reporter construct is typically transfected into a cell. After treatment with a potential modulator, the amount of reporter gene transcription, translation, or activity is WO 2005/014780 PCT/LS2004/017231 measured according to standard techniques known to those of skill in the art (Davis GF, supra; Gonzalez, J. Negulescu, P. Curr. Opin. Biotechnol. 1998: 9:624).
[0356] As outlined above, in vitro screens are done on individual genes and gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of the expression of the gene or the gene product itself is performed.
[0357] In one embodiment, screening for modulators of expression of specific gene(s) is performed. Typically, the expression of only one or a few genes is evaluated. In another embodiment, screens are designed to first find compounds that bind to differentially expressed proteins. These compounds are then evaluated for the ability to modulate differentially expressed activity. Moreover, once initial candidate compounds are identified, variants can be further screened to better evaluate structure activity relationships.
Binding Assays to Identify and Characterize Modulators [0358] In binding assays in accordance with the invention, a purified or isolated gene product of the invention is generally used. For example, antibodies are generated to a protein of the invention, and immunoassays are run to determine the amount and/or location of protein. Alternatively, cells comprising the cancer proteins are used in the assays.
[0359] Thus, the methods comprise combining a cancer protein of the invention and a candidate compound such as a ligand, and determining the binding of the compound to the cancer protein of the invention. Preferred embodiments utilize the human cancer protein; animal models of human disease of can also be developed and used. Also, other analogous mammalian proteins also can be used as appreciated by those of skill in the art. Moreover, in some embodiments variant or derivative cancer proteins are used.
[0360] Generally, the cancer protein of the invention, or theligand, is non-diffusibly bound to an insoluble support.
The support can, be one having isolated sample receiving areas (a microtiter plate, an array, etc.). The insoluble supports can be made of any composition to which the compositions can be bound, is readily separated from soluble material, and is otherwise compatible with the overall method of screening. The surface of such supports can be solid or porous and of any convenient shape.
[0361] Examples of suitable insoluble supports include microtiter plates, arrays, membranes and beads. These are typically made of glass, plastic polystyrene), polysaccharide, nylon, nitrocellulose, or TeflonTM, etc. Microtiter plates and arrays are especially convenient because a large number of assays can be carried out simultaneously, using small amounts of reagents and samples. The particular manner of binding of the composition to the support is not crucial so long as it is compatible with the reagents and overall methods of the invention, maintains the activity of the composition and is nondiffusable. Preferred methods of binding include the use of antibodies which do not sterically block either the ligand binding site or activation sequence when attaching the protein to the support, direct binding to "sticky" or ionic supports, chemical crosslinking, the synthesis of the protein or agent on the surface, etc. Following binding of the protein or ligand/binding agent to the support, excess unbound material is removed by washing. The sample receiving areas may then be blocked through incubation with bovine serum albumin (BSA), casein or other innocuous protein or other moiety.
[0362] Once a cancer protein of the invention is bound to the support, and a test compound is added to the assay.
Alternatively, the candidate binding agent is bound to the support and the cancer protein of the invention is then added.
Binding agents include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc.
WO 2005/014780 PCT/US2004/017231 [0363] Of particular interest are assays to identify agents that have a low toxicity for human cells. A wide variety of assays can be used for this purpose, including proliferation assays, cAMP assays, labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
[0364] A determination of binding of the test compound (ligand, binding agent, modulator, etc.) to a cancer protein of the invention can be done in a number of ways. The test compound can be labeled, and binding determined directly, e.g., by attaching all or a portion of the cancer protein of the invention to a solid support, adding a labeled candidate compound a fluorescent label), washing off excess reagent, and determining whether the label is present on the solid support.
Various blocking and washing steps can be utilized as appropriate.
[0365] In certain embodiments, only one of the components is labeled, a protein of the invention or ligands labeled. Alternatively, more than one component is labeled with different labels, 1125, for the proteins and a fluorophor for the compound. Proximity reagents, quenching or energy transfer reagents are also useful.
Competitive Binding to Identify and Characterize Modulators [0366] In one embodiment, the binding of the "test compound" is determined by competitive binding assay with a "competitor." The competitor is a binding moiety that binds to the target molecule a cancer protein of the invention).
Competitors include compounds such as antibodies, peptides, binding partners, ligands, etc. Under certain circumstances, the competitive binding between the test compound and the competitor displaces the test compound. In one embodiment, the test compound is labeled. Either the test compound, the competitor, or both, is added to the protein for a time sufficient to allow binding. Incubations are performed at a temperature that facilitates optimal activity, typically between four and Incubation periods are typically optimized, to facilitate rapid high throughput screening; typically between zero and one hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
[0367] In one embodiment, the competitor is added first, followed by the test compound. Displacement of the competitor is an indication that the test compound is binding to the cancer protein and thus is capable of binding to, and potentially modulating, the activity of the cancer protein. In this embodiment, either component can be labeled. Thus, e.g., if the competitor is labeled, the presence of label in the post-test compound wash solution indicates displacement by the test compound. Alternatively, if the test compound is labeled, the presence of the label on the support indicates displacement.
[0368] In an alternative embodiment, the test compound is added first, with incubation and washing, followed by the competitor. The absence of binding by the competitor indicates that the test compound binds to the cancer protein with higher affinity than the competitor. Thus, if the test compound is labeled, the presence of the label on the support, coupled with a lack of competitor binding, indicates that the test compound binds to and thus potentially modulates the cancer protein of the invention.
[0369] Accordingly, the competitive binding methods comprise differential screening to identity agents that are capable of modulating the activity of the cancer proteins of the invention. In this embodiment, the methods comprise combining a cancer protein and a competitor in a first sample. A second sample comprises a test compound, the cancer protein, and a competitor. The binding of the competitor is determined for both samples, and a change, or difference in binding between the two samples indicates the presence of an agent capable of binding to the cancer protein and WO 2005/014780 PCT/US2004/017231 potentially modulating its activity. That is, if the binding of the competitor is different in the second sample relative to the first sample, the agent is capable of binding to the cancer protein.
[0370] Alternatively, differential screening is used to identify drug candidates that bind to the native cancer protein, but cannot bind to modified cancer proteins. For example the structure of the cancer protein is modeled and used in rational drug design to synthesize agents that interact with that site, agents which generally do not bind to site-modified proteins. Moreover, such drug candidates that affect the activity of a native cancer protein are also identified by screening drugs for the ability to either enhance or reduce the activity of such proteins.
[0371] Positive controls and negative controls can be used in the assays. Preferably control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of all samples occurs for a time sufficient to allow for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined. For example, where a radiolabel is employed, the samples can be counted in a scintillation counter to determine the amount of bound compound.
[0372] A variety of other reagents can be included in the screening assays. These include reagents like salts, neutral proteins, e.g. albumin, detergents, etc. which are used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., can be used. The mixture of components is added in an order that provides for the requisite binding.
Use of Polvnucleotides to Down-requlate or Inhibit a Protein of the Invention.
[0373] Polynucleotide modulators of cancer can be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand-binding molecule, as described in WO 91/04753. Suitable ligand-binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors. Preferably, conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell. Alternatively, a polynucleotide modulator of cancer can be introduced into a cell containing the target nucleic acid sequence, by formation of a polynucleotide-lipid complex, as described in WO 90/10448. It is understood that the use of antisense molecules or knock out and knock in models may also be used in screening assays as discussed above, in addition to methods of treatment.
Inhibitory and Antisense Nucleotides [0374] In certain embodiments, the activity of a cancer-associated protein is down-regulated, or entirely inhibited, by the use of antisense polynucleotide or inhibitory small nuclear RNA (snRNA), a nucleic acid complementary to, and which can preferably hybridize specifically to, a coding mRNA nucleic acid sequence, a cancer protein of the invention, mRNA, or a subsequence thereof. Binding of the antisense polynucleotide to the mRNA reduces the translation and/or stability of the mRNA.
[0375] In the context of this invention, antisense polynucleotides can comprise naturally occurring nucleotides, or synthetic species formed from naturally occurring subunits or their close homologs. Antisense polynucleotides may also have altered sugar moieties or inter-sugar linkages. Exemplary among these are the phosphorothioate and other sulfur containing species which are known for use in the art. Analogs are comprised by this invention so long as they function WO 2005/014780 PCT/US2004/017231 effectively to hybridize with nucleotides of the invention. See, Isis Pharmaceuticals, Carlsbad, CA; Sequitor, Inc., Natick, MA.
[0376] Such antisense polynucleotides can readily be synthesized using recombinant means, or can be synthesized in vitro. Equipment for such synthesis is sold by several vendors, including Applied Biosystems. The preparation of other oligonucleotides such as phosphorothioates and alkylated derivatives is also well known to those of skill in the art.
[0377] Antisense molecules as used herein include antisense or sense oligonucleotides. Sense oligonucleotides can, be employed to block transcription by binding to the anti-sense strand. The antisense and sense oligonucleotide comprise a single stranded nucleic acid sequence (either RNA or DNA) capable of binding to target mRNA (sense) or DNA (antisense) sequences for cancer molecules. Antisense or sense oligonucleotides, according to the present invention, comprise a fragment generally at least about 12 nucleotides, preferably from about 12 to 30 nucleotides. The ability to derive an antisense or a sense oligonucleotide, based upon a cDNA sequence encoding a given protein is described in, Stein &Cohen (Cancer Res. 48:2659 (1988 and van der Krol et al. (BioTechniques 6:958 (1988)).
Ribozymes [0378] In addition to antisense polynucleotides, ribozymes can be used to target and inhibit transcription of cancerassociated nucleotide sequences. A ribozyme is an RNA molecule that catalytically cleaves other RNA molecules.
Different kinds of ribozymes have been described, including group I ribozymes, hammerhead ribozymes, hairpin ribozymes, RNase P, and axhead ribozymes (see, Castanotto et al., Adv. in Pharmacology 25: 289-317 (1994) for a general review of the properties of different ribozymes).
[0379] The general features of hairpin ribozymes are described, in Hampel et al., Nucl. Acids Res. 18:299-304 (1990); European Patent Publication No. 0360257; U.S. Patent No. 5,254,678. Methods of preparing are well known to those of skill in the art (see, WO 94/26877; Ojwang et al., Proc. Natl. Acad. Sci. USA 90:6340-6344 (1993); Yamada et al., Human Gene Therapy 1:39-45 (1994); Leavitt et al., Proc. Natl. Acad Sci. USA 92:699- 703 (1995); Leavitt et al., Human Gene Therapy 5:1151-120 (1994); and Yamada et al., Virology 205: 121-126 (1994)).
Use of Modulators in Phenotypic Screening [0380] In one embodiment, a test compound is administered to a population of cancer cells, which have an associated cancer expression profile. By "administration" or "contacting" herein is meant that the modulator is added to the cells in such a manner as to allow the modulator to act upon the cell, whether by uptake and intracellular action, or by action at the cell surface. In some embodiments, a nucleic acid encoding a proteinaceous agent a peptide) is put into a viral construct such as an adenoviral or retroviral construct, and added to the cell, such that expression of the peptide agent is accomplished, PCT US97/01019. Regulatable gene therapy systems can also be used. Once the modulator has been administered to the cells, the cells are washed if desired and are allowed to incubate under preferably physiological conditions for some period. The cells are then harvested and a new gene expression profile is generated.
Thus, cancer tissue is screened for agents that modulate, induce or suppress, the cancer phenotype. A change in at least one gene, preferably many, of the expression profile indicates that the agent has an effect on cancer activity.
Similarly, altering a biological function or a signaling pathway is indicative of modulator activity. By defining such a signature for the cancer phenotype, screens for new drugs that alter the phenotype are devised. With this approach, the drug target need not be known and need not be represented in the original gene/protein expression screening platform, nor WO 2005/014780 PCT/US2004/017231 does the level of transcript for the target protein need to change. The modulator inhibiting function will serve as a surrogate marker [0381] As outlined above, screens are done to assess genes or gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of either the expression of the gene or the gene product itself is performed.
Use of Modulators to Affect Peptides of the Invention [0382] Measurements of cancer polypeptide activity, or of the cancer phenotype are performed using a variety of assays. For example, the effects of modulators upon the function of a cancer polypeptide(s) are measured by examining parameters described above. A physiological change that affects activity is used to assess the influence of a test compound on the polypeptides of this invention. When the functional outcomes are determined using intact cells or animals, a variety of effects can be assesses such as, in the case of a cancer associated with solid tumors, tumor growth, tumor metastasis, neovascularization, hormone release, transcriptional changes to both known and uncharacterized gen'etic markers by Northern blots), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as cGNIP.
Methods of Identifying Characterizing Cancer-associated Sequences [0383] Expression of various gene sequences is correlated with cancer. Accordingly, disorders based on mutant or variant cancer genes are determined. In one embodiment, the invention provides methods for identifying cells containing variant cancer genes, determining the presence of, all or part, the sequence of at least one endogenous cancer gene in a cell. This is accomplished using any number of sequencing techniques. The invention comprises methods of identifying the cancer genotype of an individual, determining all or part of the sequence of at least one gene of the invention in the individual. This is generally done in at least one tissue of the individual, a tissue set forth in Table I, and may include the evaluation of a number of tissues or different samples of the same tissue. The method may include comparing the sequence of the sequenced gene to a known cancer gene, a wild-type gene to determine the presence of family members, homologies, mutations or variants. The sequence of all or part of the gene can then be compared to the sequence of a known cancer gene to determine If any differences exist. This is done using any number of known homology programs, such as BLAST, Bestfit, etc. The presence of a difference in the sequence between the cancer gene of the patient and the known cancer gene correlates with a disease state or a propensity for a disease state, as outlined herein.
In a preferred embodiment, the cancer genes are used as probes to determine the number of copies of the cancer gene in the genome. The cancer genes are used as probes to determine the chromosomal localization of the cancer genes.
Information such as chromosomal localization finds use in providing a diagnosis or prognosis in particular when chromosomal abnormalities such as translocations, and the like are identified in the cancer gene locus.
XIV.) Kits/Articles of Manufacture [0384] For use in the laboratory, prognostic, prophylactic, diagnostic and therapeutic applications described herein, kits are within the scope of the invention. Such kits can comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in the method, along with a label or insert comprising instructions for use, such as a use described 83 WO 2005/014780 PCT/LS2004/017231 herein. For example, the container(s) can comprise a probe that is or can be detectably labeled. Such probe can be an antibody or polynucleotide specific for a protein or a gene or message of the invention, respectively. Where the method utilizes nucleic acid hybridization to detect the target nucleic acid, the kit can also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence. Kits can comprise a container comprising a reporter, such as a biotinbinding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, fluorescent, or radioisotope label; such a reporter can be used with, a nucleic acid or antibody. The kit can include all or part of the amino acid sequences in Figure 2 or Figure 3 or analogs thereof, or a nucleic acid molecule that encodes such amino acid sequences.
[0385] The kit of the invention will typically comprise the container described above and one or more other containers associated therewith that comprise materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
[0386], A label can be present on or with the container to indicate that the composition is used for a specific therapy or non-therapeutic application, such as a prognostic, prophylactic, diagnostic or laboratory application, and can also indicate directions for either in vivo or in vitro use, such as those described herein. Directions and or other information can also be included on an insert(s) or label(s) which is included with or on the kit. The label can be on or associated with the container. A label a can be on a container when letters, numbers or other characters forming the label are molded or etched into the container itself; a label can be associated with a container when it is present within a receptacle or carrier that also holds the container, as a package insert. The label can indicate that the composition is used for diagnosing, treating, prophylaxing or prognosing a condition, such as a neoplasia of a tissue set forth in Table I.
[0387] The terms "kit" and "article of manufacture" can be used as synonyms.
[0388] In another embodiment of the invention, an article(s) of manufacture containing compositions, such as amino acid sequence(s), small molecule(s), nucleic acid sequence(s), and/or antibody(s), materials useful for the diagnosis, prognosis, prophylaxis and/or treatment of neoplasias of tissues such as those set forth in Table I is provided. The article of manufacture typically comprises at least one container and at least one label. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers can be formed from a variety of materials such as glass, metal or plastic. The container can hold amino acid sequence(s), small molecule(s), nucleic acid sequence(s), cell population(s) and/or antibody(s). In one embodiment, the container holds a polynucleotide for use in examining the mRNA expression profile of a cell, together with reagents used for this purpose. In another embodiment a container comprises an antibody, binding fragment thereof or specific binding protein for use in evaluating protein expression of282P1 G3 in cells and tissues, or for relevant laboratory, prognostic, diagnostic, prophylactic and therapeutic purposes; indications and/or directions for such uses can be included on or with such container, as can reagents and other compositions or tools used for these purposes. In another embodiment, a container comprises materials for eliciting a cellular or humoral immune response, together with associated indications and/or directions. In another embodiment, a container comprises materials for adoptive immunotherapy, such as cytotoxic T cells (CTL) or helper T cells (HTL), together with associated indications and/or directions; reagents and other compositions or tools used for such purpose can also be included.
[0389] The container can alternatively hold a composition that is effective for treating, diagnosis, prognosing or prophylaxing a condition and can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The active agents in the composition can be an antibody capable of specifically binding 282P1 G3 and modulating the function of 282P1 G3.
84 WO 2005/014780 PCT/US2004/017231 [0390] The article of manufacture can further comprise a second container comprising a pharmaceuticallyacceptable buffer, such as phosphate-buffered saline, Ringer's solution andlor dextrose solution. It can further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, stirrers, needles, syringes, and/or package inserts with indications and/or instructions for use.
EXAMPLES
[0391] Various aspects of the invention are further described and illustrated by way of the several examples that follow, none of which is intended to limit the scope of the invention.
Exam ple 1: SSH-Generated Isolation of cDNA Fragment of the PSCA Gene [0392] Intentionally Omitted Example 2: Isolation of Full Length PSCA Encoding cDNA [0393] Intentionally Omitted Example 3: Chromosomal Mapping of PSCA [0394] Intentionally Omitted Example 4 Expression Analysis of PSCA Variants in Normal Tissues and Patient Specimens [0395] Previously, PSCA, herein referred to as PSCA v.1, was identified as an antigen expressed in prostate cancer.
Its expression was detected in greater than 80% of primary prostate cancers and in the majority of prostate metastasis. It has also been shown to be expressed in bladder cancer, ovary cancer, and pancreatic cancer; these cancers are listed in Table I. By immunohistochemical analysis, PSCA has been shown to be overexpressed on the cell surface of most urothelial transitional carcinoma, and in 60% of primary pancreatic adenocarcinomas. The PSCA expression data has been reported in patent publications (PCT/US98/04664, PCT/US/28883, PCT/US00/19967) and in peer-reviewed articles (Saffran et al., Proc Natl Acad Sci U S A. 2001 Feb 27; 98(5): 2658-2663; Amara et al., Cancer Res. 2001 Jun 15; 61(12): 4660-65; Reiter et al., Proc Natl Acad Sci USA. 1998 Feb 17; 95(4): 1735-40; Argani et al., Cancer Res. 2001 Jun 1; 61(11): 4320-24).
[0396] Specific expression of different PSCA variants was studied in normal and cancer patient specimens (Figure 14 and Figure 15). Primers were designed to differentiate between PSCA v.1/v.2/v.4, PSCA v.3 and PSCA PSCA v.1/v.2/v.4 lead to a PCR product of 425 bp, PSCA v.3 leads to a PCR product of 300 bp, whereas PSCA v.5 leads to a PCR product of 910 bp in size (Figure 14A), [0397] First strand cDNA was prepared from normal bladder, brain, heart, kidney, liver, lung, prostate, spleen, skeletal muscle, testis, pancreas, colon, stomach, pools of prostate cancer, bladder cancer, kidney cancer, colon cancer, lung cancer, ovary cancer, breast cancer, cancer metastasis, and pancreas cancer (Figure 14B). Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using the variant specific primers was performed at cycles of amplification.
WO 2005/014780 PCT/LS2004/017231 [0398] Results show expression of PSCA v.5 mainly in breast cancer, cancer metastasis, and pancreas cancer, and at lower level in colon cancer and lung cancer. PSCA v.1/v.2/v.4 PCR product was detected in prostate cancer, bladder cancer, kidney cancer, colon cancer, lung cancer, ovary cancer, breast cancer, cancer metastasis, and pancreas cancer.
Amongst normal tissues, PSCA v.1/v.2/v.4 PCR product was detected only in prostate, stomach and at lower level in kidney and lung, whereas PSCA v. 5 was not detected in any normal tissue. PSCA v.3 PCR detected product was not detected in any of the samples tested.
[0399] Primers were designed to differentiate between PSCA v.4 and PSCA v.5 (Figure 15A). PSCA v.4 lead to a PCR product of 460 bp, whereas PSCA v.5 lead to a PCR product of 945 bp in size.
[0400] First strand cDNA was prepared from normal bladder, brain, heart, kidney, liver, lung, prostate, spleen, skeletal muscle, testis, pancreas, colon, stomach, pools of prostate cancer, bladder cancer, and multi-xenograft pool (prostate cancer, kidney cancer and bladder cancer xenografts) (Figure 15B). Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using the variant specific primers was performed at 30 cycles of amplification.
[0401] Results show expression of PSCA v.4 in prostate cancer, bladder cancer, and multi-xenograft pool, normal kidney and prostate. PSCA v.5 was detected only in normal prostate and bladder cancer.
[0402] The restricted expression of PSCA variants in normal tissues and the expression detected in cancer patient specimens indicate that PSCA variants are therapeutic, prognostic, laboratory, prophylactic, and diagnostic targets for human cancers.
Example 5: Transcript Variants of PSCA [0403] As used herein, the term variant includes transcript variants and single nucleotide polymorphisms (SNPs).
Transcript variants are variants of mature mRNA from the same gene which arise by alternative transcription or alternative splicing. Alternative transcripts are transcripts from the same gene but start transcription at different points. Splice variants are mRNA variants spliced differently from the same transcript. In eukaryotes, when a multi-exon gene is transcribed from genomic DNA, the initial RNA is spliced to produce functional mRNA, which has only exons and is used for translation into an amino acid sequence. Accordingly, a given gene can have zero to many alternative transcripts and each transcript can have zero to many splice variants. Each transcript variant has a unique exon makeup, and can have different coding and/or non-coding or 3' end) portions, from the original transcript. Transcript variants can code for the same, similar or different proteins, such proteins having the same or a similar function or a different function. The variant proteins can be expressed in the same tissue at the same time, in a different tissue at the same time, or in the same tissue at different times, or in a different tissue at a different time. Proteins encoded by a transcript variant can have similar or different subcellular or extracellular localizations secreted versus intracellular).
[0404] Transcript variants are identified by a variety of art-accepted methods. For example, alternative transcripts and splice variants are identified by full-length cloning, or by use of full-length transcript and EST sequences. First, all human ESTs were grouped into clusters which show direct or indirect identity with each other. Second, ESTs in the same cluster were further grouped into sub-clusters and assembled into a consensus sequence. The original gene sequence is compared to the consensus sequence(s) or other full-length sequences. Each consensus sequence is a potential splice variant for that gene. Several confirmation modalities are known in the art, such as identification of the variant by Northern analysis, full length cloning or by use of probe libraries, etc.. Even when a variant is identified that is not yet a full-length clone, that portion of the variant is very useful as a research tool, for antigen generation or for further cloning of the full-length splice variant, using techniques known in the art.
86 WO 2005/014780 PCT/US2004/017231 [0405] Moreover, computer programs are available in the art that identify transcript variants based on genomic sequences. Genomic-based transcript variant identification programs include FgenesH Salamov and V. Solovyev, "Ab initio gene finding in Drosophila genomic DNA," Genome Research. 2000 April; 10(4):516-22); Grail (URL compbio.ornl.gov/Grail-bin/EmptyGrailForm) and GenScan (URL genes.mit.edu/GENSCAN.html). For a general discussion of splice variant identification protocols see, Southan, A genomic perspective on human proteases, FEBS Lett.
2001 Jun 8; 498(2-3):214-8; de Souza, et al., Identification of human chromosome 22 transcribed sequences with ORF expressed sequence tags, Proc. Natl Acad Sci U S A. 2000 Nov 7; 97(23):12690-3.
[0406] To further confirm the parameters of a transcript variant, a variety of techniques are available in the art, such as full-length cloning, proteomic validation, PCR-based validation, and 5' RACE validation, etc. (see Proteomic Validation: Brennan, et al., Albumin banks peninsula: a new termination variant characterized by electrospray mass spectrometry, Biochem Biophys Acta. 1999 Aug 17;1433(1-2):321-6; Ferranti P, et al., Differential splicing of premessenger RNA produces multiple forms of mature caprine alpha(sl)-casein, Eur J Biochem. 1997 Oct 1;249(1):1-7. For PCR-based Validation: Wellmann S, et al., Specific reverse transcription-PCR quantification of vascular endothelial growth factor (VEGF) splice variants by LightCycler technology, Clin Chem. 2001 Apr;47(4):654-60; Jia, et al., Discovery of new human beta-defensins using a genomics-based approach, Gene. 2001 Jan 24; 263(1-2):211-8. For PCR-based and RACE Validation: Brigle, et al., Organization of the murine reduced folate carrier gene and identification of variant splice forms, Biochem Biophys Acta. 1997 Aug 7; 1353(2): 191-8).
[0407] It is known in the art that genomic regions are modulated in cancers. When the genomic region to which a gene maps is modulated in a particular cancer, the alternative transcripts or splice variants of the gene are modulated as well. Disclosed herein is that PSCA has a particular expression profile related to cancer (see, Table Alternative transcripts and splice variants of PSCA are also involved in cancers, for example in one or more of these tissues and in certain additional tissues as well. The variants thus serve as tumor-associated markers/antigens.
[0408] Using the full-length PSCA gene together with EST sequences, four additional transcript variants were identified, designated as PSCA v.2, v.3, v.4, and v.5. The boundaries of exons in the original transcript, PSCA v.1 were shown in Table LI. Schematic structures of the transcript variant nucleic acid sequences are shown in Figure 10. In Figure 10, bars with the same graphic pattern depict stretches of contiguous genetic material, the black bars designate genomic sequence found in variant 1.
[0409] Tables LIl(a) through LV(a) are set forth on a variant-by-variant basis. Lll(a) shows the nucleotide sequences of the transcript variants. Table Llll(a) shows the alignment of the transcript variants with nucleic acid sequence of PSCA v.1 (for v.2 only) or with PSCAv.2 (for all other variants). Table LIV(a) present the amino acid translation of the transcript variants for the identified reading frame orientation. Table LV(a) displays alignments of the amino acid sequence encoded by the splice variant with that of PSCA v.1.
Example 6: Single Nucleotide Polymorphisms of PSCA [0410] A Single Nucleotide Polymorphism (SNP) is a single base pair variation in a nucleotide sequence at a specific location. At any given point of the genome, there are four possible nucleotide base pairs: A/T, C/G, G/C, and TIA. As used herein, an allele is one of a series of alternative forms of a given gene, differing in DNA sequence, and affecting a product (RNA and/or protein).
[0411] A SNP that occurs on a cDNA is called a cSNP. This cSNP may change amino acids of the protein encoded by the gene and thus change the function of the protein. Some SNPs cause inherited diseases; others contribute to 87 WO 2005/014780 PCT/US2004/017231 quantitative variations in phenotype and reactions to environmental factors including diet and drugs among individuals.
Therefore, the existence of a SNP and/or combinations of alleles (called haplotypes) have many useful applications, such as diagnosis of inherited diseases, determination of drug reactions and dosage, identification of genes responsible for diseases, and analysis of the genetic relationship between individuals Nowotny, J. M. Kwon and A. M. Goate, SNP analysis to dissect human traits," Curr. Opin. Neurobiol. 2001 Oct; 11(5):637-641; M. Pirmohamed and B. K. Park, "Genetic susceptibility to adverse drug reactions," Trends Pharmacol. Sci. 2001 Jun; 22(6):298-305; J. H. Riley, C. J. Allan, E. Lai and A. Roses, "The use of single nucleotide polymorphisms in the isolation of common disease genes," Pharmacogenomics. 2000 Feb; 1(1):39-47; R. Judson, J. C. Stephens and A. Windemuth, "The predictive power of haplotypes in clinical response," Pharmacogenomics. 2000 Feb; 1(1):15-26).
[0412] SNPs are identified by a variety of art-accepted methods Bean, "The promising voyage of SNP target discovery," Am. Clin. Lab. 2001 Oct-Nov; 20(9):18-20; K. M. Weiss, "In search of human variation," Genome Res. 1998 Jul; 8(7):691-697; M. M. She, "Enabling large-scale pharmacogenetic studies by high-throughput mutation detection and genotyping technologies," Clin. Chem. 2001 Feb; 47(2):164-172). For example, SNPs are identified by sequencing DNA fragments that show polymorphism by gel-based methods such as restriction fragment length polymorphism (RFLP) and denaturing gradient gel electrophoresis (DGGE). They are also discovered by direct sequencing of DNA samples pooled from different individuals or by comparing sequences from different DNA samples. With the rapid accumulation of sequence data in public and private databases, one also discovers SNPs by comparing sequences using computer programs Gu, L. Hillier and P. Y. Kwok, "Single nucleotide polymorphism hunting in cyberspace," Hum. Mutat. 1998; 12(4):221-225).
SNPs can be verified and the genotype or haplotype of an individual can be determined by a variety of methods including direct sequencing and high throughput microarrays Y. Kwok, "Methods for genotyping single nucleotide polymorphisms," Annu. Rev. Genomics Hum. Genet. 2001; 2:235-258; M. Kokoris, K. Dix, K. Moynihan, J. Mathis, B. Erwin, P. Grass, B. Hines and A. Duesterhoeft, "High-throughput SNP genotyping with the Masscode system," Mol. Diagn. 2000 Dec; 5(4):329-340).
[0413] Using the methods described above, thirteen SNP were identified in the transcript for PSCA v.2. Variant 2 was used, rather than for example variant 1, as it had fewer ambiguous bases than variant 1. Accordingly, SNPs were identified in PSCA v.2, at positions 57 367 424 495 499 563 567 (gla), 627 (gla), 634 835 (gla), 847 878 (gla), and 978 The transcripts or proteins with alternative alleles were designated as variant PSCA v.6 through v.18, as shown in Table LVI and Figure 12a.
[0414] The nucleotide change in v.6 changed the start codon of v.1 and thus, the translation would not start until the next ATG (AUG in mRNA), resulting in a protein 9 AA shorter than v.1 protein (Figure 11a). The nucleotide changes for v.7 and v.8 were silent at the protein level.
[0415] Twelve of these 13 SNPs were also present in variant 4 as set forth in Figure 12b and table LVI. The 12 SNP variants relative to PSCA v. 4 are designated PSCA v. 19 through v.30. Variants 19 through 27 encode alternative amino acids. (Figure 11b and Table LVI).
[0416] Table LVI also shows the amino acid changes of protein sequence. These SNP, though shown individually separately here, can occur in different combinations and in any one of the transcript variants that contains the site of the
SNP.
WO 2005/014780 PCT/US2004/017231 Example 7: Production of Recombinant PSCA in Prokarvotic Systems [0417] To express recombinant PSCA and PSCA variants in prokaryotic cells, the full or partial length PSCA and PSCA variant cDNA sequences are cloned into any one of a variety of expression vectors known in the art. One or more of the following regions of PSCA variants are expressed: the full length sequence presented in Figures 2 and 3, or any 8, 9, 11, 12, 13,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more contiguous amino acids from PSCA, variants, or analogs thereof.
A. In vitro transcription and translation constructs: [0418] gCRII: To generate PSCA sense and anti-sense RNA probes for RNA in situ investigations, pCRII constructs (Invitrogen, Carlsbad CA) are generated encoding either all or fragments of the PSCA cDNA. The pCRII vector has Sp6 and T7 promoters flanking the insert to drive the transcription of PSCA RNA for use as probes in RNA in situ hybridization experiments. These probes are used to analyze the cell and tissue expression of PSCA at the RNA level. Transcribed.
PSCA RNA representing the cDNA amino acid coding region of the PSCA gene is used in in vitro translation systems such as the TnTF M Coupled Reticulolysate System (Promega, Corp., Madison, WI) to synthesize PSCA protein.
B. Bacterial Constructs: [0419] pGEX Constructs: To generate recombinant PSCA proteins in bacteria that are fused to the Glutathione Stransferase (GST) protein, all or parts of the PSCA cDNA protein coding sequence are cloned into the pGEX family of GSTfusion vectors (Amersham Pharmacia Biotech, Piscataway, NJ). These constructs allow controlled expression of recombinant PSCA protein sequences with GST fused at the amino-terminus and a six histidine epitope (6X His) at the carboxyl-terminus. The GST and 6X His tags permit purification of the recombinant fusion protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-GST and anti-His antibodies. The 6X His tag is generated by adding 6 histidine codons to the cloning primer at the 3' end, of the open reading frame (ORF).
A proteolytic cleavage site, such as the PreScissionTM recognition site in pGEX-6P-1, may be employed such that it permits cleavage of the GST tag from PSCA-related protein. The ampicillin resistance gene and pBR322 origin permits selection and maintenance of the pGEX plasmids in E. coli.
[0420] pMAL Constructs: To generate, in bacteria, recombinant PSCA proteins that are fused to maltose-binding protein (MBP), all or parts of the PSCA cDNA protein coding sequence are fused to the MBP gene by cloning into the pMAL-c2X and pMAL-p2X vectors (New England Biolabs, Beverly, MA). These constructs allow controlled expression of recombinant PSCA protein sequences with MBP fused at the amino-terminus and a 6X His epitope tag at the carboxylterminus. The MBP and BX His tags permit purification of the recombinant protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-MBP and anti-His antibodies. The 6X His epitope tag is generated by adding 6 histidine codons to the 3' cloning primer. A Factor Xa recognition site permits cleavage of the pMAL tag from PSCA, The pMAL-c2X and pMAL-p2X vectors are optimized to express the recombinant protein in the cytoplasm or periplasm respectively. Periplasm expression enhances folding of proteins with disulfide bonds.
[0421] pET Constructs: To express PSCA in bacterial cells, all or parts of the PSCA cDNA protein coding sequence are cloned into the pET family of vectors (Novagen, Madison, WI). These vectors allow tightly controlled expression of recombinant PSCA protein in bacteria with and without fusion to proteins that enhance solubility, such as NusA and thioredoxin (Trx), and epitope tags, such as 6X His and S-Tag
T
that aid purification and detection of the recombinant WO 2005/014780 PCT/US2004/017231 protein. For example, constructs are made utilizing pET NusA fusion system 43.1 such that regions of the PSCA protein are expressed as amino-terminal fusions to NusA.
C. Yeast Constructs: [0422] pESC Constructs: To express PSCA in the yeast species Saccharomyces cerevisiae for generation of recombinant protein and functional studies, all or parts of the PSCA cDNA protein coding sequence are cloned into the pESC family of vectors each of which contain 1 of 4 selectable markers, HIS3, TRP1, LEU2, and URA3 (Stratagene, La Jolla, CA). These vectors allow controlled expression from the same plasmid of up to 2 different genes or cloned sequences containing either FlagTM or Myc epitope tags in the same yeast cell. This system is useful to confirm proteinprotein interactions of PSCA. In addition, expression in yeast yields similar post-translational modifications, such as glycosylations and phosphorylations, that are found when expressed in eukaryotic cells.
[0423] pESP Constructs: To express PSCA in the yeast species Saccharomyces pombe, all or parts of the PSCA cDNA protein coding sequence are cloned into the pESP family of vectors. These vectors allow controlled high level of expression of a PSCA protein sequence that is fused at either the amino terminus or at the carboxyl terminus to GST which aids purification of the recombinant protein. A FlagT epitope tag allows detection of the recombinant protein with anti- FlagTM antibody.
Example 8: Production of Recombinant PSCA in Higher Eukaryotic Systems [0424] Intentionally Omitted Example 9: Antigenicity Profiles and Secondary Structure [0425] Figure 5A-C, Figure 6A-C, Figure 7A-C, Figure 8A-C, and Figure 9A-C depict graphically five amino acid profiles of PSCA variants 1, 3, and 4, each assessment available by accessing the ProtScale website (URL www.expasy.ch/cgi-bin/protscale.pl) on the ExPasy molecular biology server.
[0426] These profiles: Figure 5, Hydrophilicity, (Hopp Woods 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824- 3828); Figure 6, Hydropathicity, (Kyte Doolittle 1982. J. Mol. Biol. 157:105-132); Figure 7, Percentage Accessible Residues (Janin 1979 Nature 277:491-492); Figure 8, Average Flexibility, (Bhaskaran and Ponnuswamy 1988.
Int. J. Pept. Protein Res. 32:242-255); Figure 9, Beta-turn (Deleage, Roux B. 1987 Protein Engineering 1:289-294); and optionally others available in the art, such as on the ProtScale website, were used to identify antigenic regions of each of the PSCA variant proteins. Each of the above amino acid profiles of PSCA variants were generated using the following ProtScale parameters for analysis: 1) A window size of 9; 2) 100% weight of the window edges compared to the window center; and, 3) amino acid profile values normalized to lie between 0 and 1.
[0427] Hydrophilicity (Figure Hydropathicity (Figure 6) and Percentage Accessible Residues (Figure 7) profiles were used to determine stretches of hydrophilic amino acids values greater than 0.5 on the Hydrophilicity and Percentage Accessible Residues profile, and values less than 0.5 on the Hydropathicity profile). Such regions are likely to be exposed to the aqueous environment, be present on the surface of the protein, and thus available for immune recognition, such as by antibodies.
[0428] Average Flexibility (Figure 8) and Beta-turn (Figure 9) profiles determine stretches of amino acids values greater than 0.5 on the Beta-turn profile and the Average Flexibility profile) that are not constrained in secondary structures WO 2005/014780 PCT/US2004/017231 such as beta sheets and alpha helices. Such regions are also more likely to be exposed on the protein and thus accessible to immune recognition, such as by antibodies.
[0429] Antigenic sequences of the PSCA variant proteins indicated, by the profiles set forth in Figure Figure 6A-C, Figure 7A-C, Figure 8A-C, and/or Figure 9A-C are used to prepare immunogens, either peptides or nucleic acids that encode them, to generate therapeutic and diagnostic anti-PSCA antibodies. The immunogen can be any 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50 or more than 50 contiguous amino acids, or the corresponding nucleic acids that encode them, from the PSCA protein variants listed in Figures 2 and 3 of which the amino acid profiles are shown in Figure 9, or can be inferred because the variant contains sequence that is the same as a variant depicted in figure 9. In particular, peptide immunogens of the invention can comprise, a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profiles of Figure 5; a peptide region of at least 5 amino adds of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figures 6; a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profiles of Figure 7; a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profiles on Figure 8; and, a peptide region of at least amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of Figures 9 Peptide immunogens of the invention can also comprise nucleic acids that encode any of the forgoing.
[0430] All immunogens of the invention, peptide or nucleic acid, can be embodied in human unit dose form, or comprised by a composition that includes a pharmaceutical excipient compatible with human physiology.
[0431] The secondary structure of PSCA protein variants 1, 3, 4, and 6, namely the predicted presence and location of alpha helices, extended strands, and random coils, is predicted from the primary amino acid sequence using the HNN Hierarchical Neural Network method (NPS@: Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet Blanchet Geourjon C. and DelBage http://pbil.ibcp.fr/cgibin/npsa_automat.pl?page=npsann.html), accessed from the ExPasy molecular biology server (http://www.expasy.ch/tools/). The analysis indicates that PSCA variant 1 is composed of 30.89% alpha helix, 21.95% extended strand, and 47.15% random coil (Figure 13A). PSCA protein variant 3 is composed of 14.89% alpha helix, 8.51% extended strand, and 76.60% random coil (Figure 13B). PSCA protein variant 4 is composed of 9.52% alpha helix, 8.99% extended strand, and 81.48% random coil (Figure 13C). PSCA protein variant 6 is composed of 24.56% alpha helix, 21.93% extended strand, and 53.51% random coil (Figure 13D).
[0432] Analysis for the potential presence of transmembrane domains in the PSCA variant proteins was carried out using a variety of transmembrane prediction algorithms accessed from the ExPasy molecular biology server (http://www.expasy.ch/tools/). Shown graphically in figure 13E, G, I, and K are the results of analyses of variants 1, 3, 4, and 6, respectively, using the TMpred program. Shown graphically in figure 13F, H, J, and L are the results of analyses of variants 1, 3, 4, and 6, respectively using the TMHMM program. PSCA variant 1 and variant 6 proteins are likely to encode GPI-linked proteins. Variants 3 and 4 are likely to encode soluble proteins since they do not contain significant predictions for transmembrane domains. The results of structural analysis programs are summarized in Table VI.
WO 2005/014780 PCT/US2004/017231 Example 10: Generation of PSCA Polyclonal Antibodies [0433] Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant. Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections. In addition to immunizing with a full length PSCA protein variant, computer algorithms are employed in design of immunogens that, based on amino acid sequence analysis contain characteristics of being antigenic and available for recognition by'the immune system of the immunized host (see the Example entitled "Antigenicity Profiles and Secondary Structure"). Such regions would be predicted to be hydrophilic, flexible, in beta-turn conformations, and be exposed on the surface of the protein (see, Figure 5A-C, Figure 6A-C, Figure 7A-C, Figure 8A-C, or Figure 9A-C for amino acid profiles that indicate such regions of PSCA protein variant 1).
[0434] For example, recombinant bacterial fusion proteins or peptides containing hydrophilic, flexible, beta-turn regions of PSCA protein variants are used as antigens to generate polyclonal antibodies in New Zealand White rabbits or monoclonal antibodies as described in Example 11. For example, in PSCA variant 1, such regions include, but are not limited to, amino acids 28-56 and amino acids 66-94. For variant 3, such regions include, but are not limited to, amino acids 7-39 and amino acids 70-94. For variant 4 such regions include, but are not limited to, amino acids 6-18, amino acids 27-39, amino acids 103-133, and 177-189. For variant 6, such regions include, but are not limited to, amino acids 19-35 and amino acids 57-85. It is useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include, but are not limited to, keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. In one embodiment, a peptide encoding amino acids 103-133 of PSCA variant 4 is conjugated to KLH and used to immunize a rabbit. Alternatively the immunizing agent may include all or portions of the PSCA variant proteins, analogs or fusion proteins thereof. For example, the PSCA variants amino acid sequences can be fused using recombinant DNA techniques to any one of a variety of fusion protein partners that are well known in the art, such as glutathione-S-transferase (GST) and HIS tagged fusion proteins. In one embodiment, the PSCA variant 1 sequence, amino acids 18-98 was fused to GST using recombinant techniques in the pGEX expression vector, expressed, purified and used to immunize both rabbits and mice to generate polyclonal and monoclonal antibodies respectively. Such fusion proteins are purified from induced bacteria using the appropriate affinity matrix.
[0435] Other recombinant bacterial fusion proteins that may be employed include maltose binding protein, LacZ, thioredoxin, NusA, or an immunoglobulin constant region (see the section entitled "Production of PSCA in Prokaryotic Systems" and Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubul et al. eds., 1995; Linsley, Brady, Urnes, Grosmaire, L, Damle, and Ledbetter, L.(1991) J.Exp. Med. 174, 561-566).
[0436] In addition to bacterial derived fusion proteins, mammalian expressed protein antigens are also used. These antigens are expressed from mammalian expression vectors such as the Tag5 and Fc-fusion vectors (see the section entitled "Production of Recombinant PSCA in Eukaryotic Systems"), and retain post-translational modifications such as glycosylations found in native protein. In one embodiment, the cDNA of PSCA variant 1, minus the N-terminal leader peptide and C-terminal GPI anchor was cloned into the Tag5 mammalian secretion vector, and expressed in 293T cells.
The recombinant protein was purified by metal chelate chromatography from tissue culture supernatants of 293T cells stably expressing the recombinant vector. The purified Tag5 PSCA protein was then used as immunogen.
[0437] During the immunization protocol, it is useful to mix or emulsify the antigen in adjuvants that enhance the immune response of the host animal. Examples of adjuvants include, but are not limited to, complete Freund's adjuvant (CFA) and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
92 WO 2005/014780 PCT/US2004/017231 [0438] In a typical protocol, rabbits are initially immunized subcutaneously with up to 200 jig, typically 100-200 jg, of fusion protein or peptide conjugated to KLH mixed in complete Freund's adjuvant (CFA). Rabbits are then injected subcutaneously every two weeks with up to 200 j.g, typically 100-200 gg, of the immunogen in incomplete Freund's adjuvant (IFA). Test bleeds are taken approximately 7-10 days following each immunization and used to monitor the titer of the antiserum by ELISA.
[0439] To test reactivity and specificity of immune serum, such as rabbit serum derived from immunization with a GST-fusion of PSCA variant 3 or 4 protein, the respective full-length PSCA variant cDNA is cloned into pCDNA 3.1 myc-his expression vector (Invitrogen, see the Example entitled "Production of Recombinant PSCA in Eukaryotic Systems"). After transfection of the constructs into 293T cells, cell lysates are probed with the anti-variant serum and with anti-His antibody (Santa Cruz Biotechnologies, Santa Cruz, CA) to determine specific reactivity to denatured variant protein using the Western blot technique. In addition, the immune serum is tested by fluorescence microscopy, flow cytometry and immunoprecipitation against 293T and other recombinant PSCA variant-expressing cells to determine specific recognition of native protein. Westem blot, immunoprecipitation, fluorescent microscopy, and flow cytometric techniques using cells that endogenously express PSCA are also carried out to test reactivity and specificity.
[0440] Anti-serum from rabbits immunized with PSCA variant fusion proteins, such as GST and MBP fusion proteins, are purified by depletion of antibodies reactive to the fusion partner sequence by passage over an affinity column containing the fusion partner either alone or in the context of an irrelevant fusion protein. For example, antiserum derived from a GST-PSCA variant 1 fusion protein is first purified by passage over a column of GST protein covalently coupled to AffiGel matrix (BioRad, Hercules, Calif.). The antiserum is then affinity purified by passage over a column composed of a MBP-PSCA fusion protein covalently coupled to Affigel matrix. The serum is then further purified by protein G affinity chromatography to isolate the IgG fraction. Sera from other His-tagged antigens and peptide immunized rabbits as well as fusion partner depleted sera are affinity purified by passage over a column matrix composed of the original protein immunogen or free peptide.
Example 11: Generation of PSCA Monoclonal Antibodies (mAbs) [0441] In one embodiment, therapeutic mAbs to PSCA variants comprise those that react with epitopes specific for each variant protein or specific to sequences in common between the variants that would disrupt or modulate the biological function of the PSCA variants, for example those that would disrupt the interaction with ligands and binding partners.
Immunogens for generation of such mAbs include those designed to encode or contain the entire PSCA protein variant sequence, regions of the PSCA protein variants predicted to be antigenic from computer analysis of the amino acid sequence (see, Figure 5A-C, Figure 6A-C, Figure 7A-C, Figure 8A-C, or Figure 9A-C, and the Example entitled "Antigenicity Profiles"). Immunogens include peptides, recombinant bacterial proteins, and mammalian expressed Tag proteins and human and murine IgG FC fusion proteins. In addition, cells engineered to express high levels of a respective PSCA variant, such as 293T-PSCA variant 4 or 300.19-PSCA variant 4 murine Pre-B cells, are used to immunize mice.
[0442] To generate mAbs to a PSCA variant, mice are first immunized intraperitoneally (IP) with, typically, 10-50 4g of protein immunogen or 107 PSCA-expressing cells mixed in complete Freund's adjuvant. Mice are then subsequently immunized IP every 2-4 weeks with, typically, 10-50 ig of protein immunogen or 107 cells mixed in incomplete Freund's adjuvant. Alternatively, MPL-TDM adjuvant is used in immunizations. In addition to the above protein and cell-based immunization strategies, a DNA-based immunization protocol is employed in which a mammalian expression vector WO 2005/014780 PCT/US2004/017231 encoding a PSCA variant sequence is used to immunize mice by direct injection of the plasmid DNA. For example, the complete cDNA of PSCA of variant 4 is cloned into the Tag5 mammalian secretion vector and the recombinant vector will then be used as immunogen. In another example the same amino acids are cloned into an Fc-fusion secretion vector in which the PSCA variant 4 sequence is fused at the amino-terminus to an IgK leader sequence and at the carboxyl-terminus to the coding sequence of the human or murine IgG Fc region. This recombinant vector is then used as immunogen. The plasmid immunization protocols are used in combination with purified proteins expressed from the same vector and with cells expressing the respective PSCA variant.
[0443] During the immunization protocol, test bleeds are taken 7-10 days following an injection to monitor titer and specificity of the immune response. Once appropriate reactivity and specificity is obtained as determined by ELISA, Western blotting, immunoprecipitation, fluorescence microscopy, and flow cytometric analyses, fusion and hybridoma generation is then carried out with established procedures well known in the art (see, Harlow and Lane, 1988).
[0444] In one embodiment for generating PSCA monoclonal antibodies, a GST-fusion of variant 4 antigen encoding amino acids 1-189 is expressed and then purified from stably transfected 293T cells. Balb C mice are initially immunized intraperitoneally with 25 tg of the GST-PSCA variant 4 protein mixed in complete Freund's adjuvant. Mice are subsequently immunized every two weeks with 25 tg of the antigen mixed in incomplete Freund's adjuvant for a total of three immunizations. ELISA using the GST-fusion antigen and a cleavage product from which the GST portion is removed determines the titer of serum from immunized mice. Reactivity and specificity of serum to full length PSCA variant 4 protein is monitored by Western blotting, immunoprecipitation and flow cytometry using 293T cells transfected with an expression vector encoding the PSCA variant 1 cDNA (see the Example entitled "Production of Recombinant PSCA in Eukaryotic Systems"). Other recombinant PSCA variant 4-expressing cells or cells endogenously expressing PSCA variant 4 are also used. Mice showing the strongest reactivity are rested and given a final injection of Tag5 antigen in PBS and then sacrificed four days later. The spleens of the sacrificed mice are harvested and fused to SPO2 myeloma cells using standard procedures (Harlow and Lane, 1988). Supernatants from HAT selected growth wells are screened by ELISA, Western blot, immunoprecipitation, fluorescent microscopy, and flow cytometry to identify PSCA specific antibodyproducing clones.
[0445] To generate monoclonal antibodies that are specific for PSCA variant 4 protein, immunogens are designed to encode the sequence unique to that variant. For example, a peptide encoding amino acids 6-18 of PSCA variant 4 is synthesized, conjugated to KLH and used as immunogen. Hybridoma supernatants are then screened on the peptide antigen and then further screened on cells expressing the PSCA variant 4 protein and cross-screened on cells expressing the other PSCA variants to derive variant 4-specific monoclonal antibodies.
[0446] The binding affinity of a PSCA variant monoclonal antibody is determined using standard technologies.
Affinity measurements quantify the strength of antibody to epitope binding and are used to help define which PSCA variant monoclonal antibodies preferred for diagnostic or therapeutic use, as appreciated by one of skill in the art. The BIAcore system (Uppsala, Sweden) is a preferred method for determining binding affinity. The BlAcore system uses surface plasmon resonance (SPR, Welford K. 1991, Opt. Quant. Elect. 23:1; Morton and Myszka, 1998, Methods in Enzymology 295: 268) to monitor biomolecular interactions in real time. BIAcore analysis conveniently generates association rate constants, dissociation rate constants, equilibrium dissociation constants, and affinity constants.
WO 2005/014780 PCT/US2004/017231 Example 12: HLA Class I and Class II Binding Assays HLA class I and class II binding assays using purified HLA molecules are performed in accordance with disclosed protocols PCT publications WO 94/20127 and WO 94/03205; Sidney et al., Current Protocols in Immunology 18.3.1 (1998); Sidney, et J. Immunol. 154:247 (1995); Sette, et al., Mol. Immunol. 31:813 (1994)). Briefly, purified MHC molecules (5 to 500 nM) are incubated with various unlabeled peptide inhibitors and 1-10 nM 1 2 5 1-radiolabeled probe peptides as described. Following incubation, MHC-peptide complexes are separated from free peptide by gel filtration and the fraction of peptide bound is determined. Typically, in preliminary experiments, each MHC preparation is titered in the presence of fixed amounts of radiolabeled peptides to determine the concentration of HLA molecules necessary to bind of the total radioactivity. All subsequent inhibition and direct binding assays are performed using these HLA concentrations.
Since under these conditions [label]<[HLA] and ICso>[HLA], the measured ICso values are reasonable approximations of the true KD values. Peptide inhibitors are typically tested at concentrations ranging from 120 ig/ml to 1.2 ng/ml, and are tested in two to four completely independent experiments. To allow comparison of the data obtained in different experiments, a relative binding figure is calculated for each peptide by dividing the ICso of a positive control for inhibition by the ICso for each tested peptide (typically unlabeled versions of the radiolabeled probe peptide). For database purposes, and inter-experiment comparisons, relative binding values are compiled. These values can subsequently be converted back into ICso nM values by dividing the ICso nM of the positive controls for inhibition by the relative binding of the peptide of interest. This method of data compilation is accurate and consistent for comparing peptides that have been tested on different days, or with different lots of purified MHC.
Binding assays as outlined above may be used to analyze HLA supermotif and/or HLA motif-bearing peptides (see Table IV).
Example 13: Identification of HLA Supermotif- and Motif-Bearing CTL Candidate Epitopes [0447] HLA vaccine compositions of the invention can include multiple epitopes. The multiple epitopes can comprise multiple HLA supermotifs or motifs to achieve broad population coverage. This example illustrates the identification and confirmation of supermotif- and motif-bearing epitopes for the inclusion in such a vaccine composition. Calculation of population coverage is performed using the strategy described below.
Computer searches and algorithms for identification of supermotif and/or motif-bearing eoitopes [0448] The searches performed to identify the motif-bearing peptide sequences in the Example entitled "Antigenicity Profiles" and Tables VIII-XXI and XXII-XLIX employ the protein sequence data from the gene product of PSCA set forth in Figures 2 and 3, the specific search peptides used to generate the tables are listed in Table VII.
[0449] Computer searches for epitopes bearing HLA Class I or Class II supermotifs or motifs are performed as follows. All translated PSCA protein sequences are analyzed using a text string search software program to identify potential peptide sequences containing appropriate HLA binding motifs; such programs are readily produced in accordance with information in the art in view of known motif/supermotif disclosures. Furthermore, such calculations can be made mentally.
WO 2005/014780 PCT/US2004/017231 [0450] Identified A2-, A3-, and DR-supermotif sequences are scored using polynomial algorithms to predict their capacity to bind to specific HLA-Class I or Class II molecules. These polynomial algorithms account for the impact of different amino acids at different positions, and are essentially based on the premise that the overall affinity (or AG) of peptide-HLA molecule interactions can be approximated as a linear polynomial function of the type: "AG" ali x a2 x a3i x an where aji is a coefficient which represents the effect of the presence of a given amino acid at a given position along the sequence of a peptide of n amino acids. The crucial assumption of this method is that the effects at each position are essentially independent of each other independent binding of individual side-chains). When residuej occurs at position i in the peptide, it is assumed to contribute a constant amountji to the free energy of binding of the peptide irrespective of the sequence of the rest of the peptide.
[0451] The method of derivation of specific algorithm coefficients has been described in Gulukota et J. Mol. Biol.
267:1258-126,1997; (see also Sidney et al., Human Immunol. 45:79-93, 1996; and Southwood et J. Immunol.
160:3363-3373, 1998). Briefly, for all i positions, anchor and non-anchor alike, the geometric mean of the average relative binding (ARB) of all peptides carrying j is calculated relative to the remainder of the group, and used as the estimate ofji.
For Class II peptides, if multiple alignments are possible, only the highest scoring alignment is utilized, following an iterative procedure. To calculate an algorithm score of a given peptide in a test set, the ARB values corresponding to the sequence of the peptide are multiplied. If this product exceeds a chosen threshold, the peptide is predicted to bind. Appropriate thresholds are chosen as a function of the degree of stringency of prediction desired.
Selection of HLA-A2 supertype cross-reactive peptides [0452] Protein sequences from PSCA are scanned utilizing motif identification software, to identify 9- 10- and 11mer sequences containing the HLA-A2-supermotif main anchor specificity. Typically, these sequences are then scored using the protocol described above and the peptides corresponding to the positive-scoring sequences are synthesized and tested for their capacity to bind purified HLA-A*0201 molecules in vitro (HLA-A*0201 is considered a prototype A2 supertype molecule).
[0453] These peptides are then tested for the capacity to bind to additional A2-supertype molecules (A*0202, A*0203, A*0206, and A*6802). Peptides that bind to at least three of the five A2-supertype alleles tested are typically deemed A2-supertype cross-reactive binders. Preferred peptides bind at an affinity equal to or less than 500 nM to three or more HLA-A2 supertype molecules.
Selection of HLA-A3 supermotif-bearing epitopes [0454] The PSCA protein sequence(s) scanned above is also examined for the presence of peptides with the HLA- A3-supermotif primary anchors. Peptides corresponding to the HLA A3 supermotif-bearing sequences are then synthesized and tested for binding to HLA-A*0301 and HLA-A*1101 molecules, the molecules encoded by the two most prevalent A3-supertype alleles. The peptides that bind at least one of the two alleles with binding affinities of 500 nM, often 200 nM, are then tested for binding cross-reactivity to the other common A3-supertype alleles A*3101, A*3301, and A*6801) to identify those that can bind at least three of the five HLA-A3-supertype molecules tested.
WO 2005/014780 PCT/US2004/017231 Selection of HLA-B7 supermotif bearing epitopes [0455] The PSCA protein(s) scanned above is also analyzed for the presence of 9-10-, or 11-mer peptides with the HLA-B7-supermotif. Corresponding peptides are synthesized and tested for binding to HLA-B*0702, the molecule encoded by the most common B7-supertype allele the prototype B7 supertype allele). Peptides binding B*0702 with of_ 500 nM are identified using standard methods. These peptides are then tested for binding to other common B7supertype molecules B*3501, B*5101, B*5301, and B*5401). Peptides capable of binding to three or more of the five B7-supertype alleles tested are thereby identified.
Selection of Al and A24 motif-bearing epitopes [0456] To further increase population coverage, HLA-A1 and -A24 epitopes can also be incorporated into vaccine compositions. An analysis of the PSCA protein can also be performed to identify HLA-A1- and A24-motif-containing sequences.
[0457] High affinity and/or cross-reactive binding epitopes that bear other motif and/or supermotifs are identified using analogous methodology.
Example 14: Confirmation of Immunoqenicity [0458] Cross-reactive candidate CTL A2-supermotif-bearing peptides that are identified as described herein are selected to confirm in vitro immunogenicity. Confirmation is performed using the following methodology: Target Cell Lines for Cellular Screeninq: [0459] The .221A2.1 cell line, produced by transferring the HLA-A2.1 gene into the HLA-A, -C null mutant human B-lymphoblastoid cell line 721.221, is used as the peptide-loaded target to measure activity of HLA-A2.1-restricted CTL.
This cell line is grown in RPMI-1640 medium supplemented with antibiotics, sodium pyruvate, nonessential amino acids and 10% heat inactivated FCS. Cells that express an antigen of interest, or transfectants comprising the gene encoding the antigen of interest, can be used as target cells to confirm the ability of peptide-specific CTLs to recognize endogenous antigen.
Primary CTL Induction Cultures: [0460] Generation of Dendritic Cells PBMCs are thawed in RPMI with 30 g/ml DNAse, washed twice and resuspended in complete medium (RPMI-1640 plus 5% AB human serum, non-essential amino acids, sodium pyruvate, Lglutamine and penicillin/streptomycin). The monocytes are purified by plating 10 x 106 PBMC/well in a 6-well plate. After 2 hours at 37°C, the non-adherent cells are removed by gently shaking the plates and aspirating the supernatants. The wells are washed a total of three times with 3 ml RPMI to remove most of the non-adherent and loosely adherent cells. Three ml of complete medium containing 50 ng/ml of GM-CSF and 1,000 Ulml of IL-4 are then added to each well. TNFa is added to the DCs on day 6 at 75 ng/ml and the cells are used for CTL induction cultures on day 7.
[0461] Induction of CTL with DC and Peptide: CD8+ T-cells are isolated by positive selection with Dynal immunomagnetic beads (Dynabeads@ M-450) and the detacha-bead® reagent. Typically about 200-250x106 PBMC are processed to obtain 24x10 6 CD8+ T-cells (enough for a 48-well plate culture). Briefly, the PBMCs are thawed in RPMI with 304g/ml DNAse, washed once with PBS containing 1% human AB serum and resuspended in PBS/1% AB serum at a WO 2005/014780 PCT/US2004/017231 concentration of 20x10 6 cells/ml. The magnetic beads are washed 3 times with PBS/AB serum, added to the cells (140pl beads/20x105 cells) and incubated for 1 hour at 4°C with continuous mixing. The beads and cells are washed 4x with PBS/AB serum to remove the nonadherent cells and resuspended at 100x10 6 cells/ml (based on the original cell number) in PBS/AB serum containing 100Il/ml detacha-bead® reagent and 30 pg/ml DNAse. The mixture is incubated for 1 hour at room temperature with continuous mixing. The beads are washed again with PBS/AB/DNAse to collect the CD8+ T-cells.
The DC are collected and centrifuged at 1300 rpm for 5-7 minutes, washed once with PBS with 1% BSA, counted and pulsed with 40pg/ml of peptide at a cell concentration of 1-2x10 6 /ml in the presence of 3pg/ml 112- microglobulin for 4 hours at 20°C. The DC are then irradiated (4,200 rads), washed 1 time with medium and counted again.
[0462] Setting up induction cultures: 0.25 ml cytokine-generated DC (at lx105 cells/ml) are co-cultured with 0.25ml of CD8+ T-cells (at 2x106 cell/ml) in each well of a 48-well plate in the presence of 10 ng/ml of IL-7. Recombinant human is added the next day at a final concentration of 10 ng/ml and rhuman IL-2 is added 48 hours later at 10 IU/ml.
[0463] Restimulation of the induction cultures with peptide-pulsed adherent cells: Seven and fourteen days after the primary induction, the cells are restimulated with peptide-pulsed adherent cells. The PBMCs are thawed and washed twice with RPMI and DNAse. The cells are resuspended at 5x106 cells/ml and irradiated at-4200 rads. The PBMCs are plated at 2x106 in 0.5 ml complete medium per well and incubated for 2 hours at 370C. The plates are washed twice with RPMI by tapping the plate gently to remove the nonadherent cells and the adherent cells pulsed with 10pg/ml of peptide in the presence of 3 pg/mI 1R2 microglobulin in 0.25ml RPMV5%AB per well for 2 hours at 37°C. Peptide solution from each well is aspirated and the wells are washed once with RPMI. Most of the media is aspirated from the induction cultures (CD8+ cells) and brought to 0,5 ml with fresh media. The cells are then transferred to the wells containing the peptide-pulsed adherent cells. Twenty four hours later recombinant human IL-10 is added at a final concentration of 10 ng/ml and recombinant human IL2 is added the next day and again 2-3 days later at 501U/ml (Tsai et al., Critical Reviews in immunology 18(1-2):65-75, 1998). Seven days later, the cultures are assayed for CTL activity in a 51 Cr release assay. In some experiments the cultures are assayed for peptide-specific recognition in the in situ IFNy ELISA at the time of the second restimulation followed by assay of endogenous recognition 7 days later. After expansion, activity is measured in both assays for a side-by-side comparison.
Measurement of CTL lvtic activity by 51 Cr release.
[0464] Seven days after the second restimulation, cytotoxicity is determined in a standard (5 hr) 51 Cr release assay by assaying individual wells at a single E:T. Peptide-pulsed targets are prepared by incubating the cells with peptide overnight at 37°C.
[0465] Adherent target cells are removed from culture flasks with trypsin-EDTA. Target cells are labeled with 200pCi of 5 1Cr sodium chromate (Dupont, Wilmington, DE) for 1 hour at 370C. Labeled target cells are resuspended at 106 per ml and diluted 1:10 with K562 cells at a concentration of 3.3x10 6 /ml (an NK-sensitive erythroblastoma cell line used to reduce non-specific lysis). Target cells (100 pl) and effectors (100pl) are plated in 96 well round-bottom plates and incubated for 5 hours at 37°C. At that time, 100 pi of supernatant are collected from each well and percent lysis is determined according to the formula: [(cpm of the test sample- cpm of the spontaneous 5 1Cr release sample)/(cpm of the maximal 6 Cr release samplecpm of the spontaneous 51 Cr release sample)] x 100.
[0466] Maximum and spontaneous release are determined by incubating the labeled targets with 1% Triton X-100 and media alone, respectively. A positive culture is defined as one in which the specific lysis (sample- background) is 98 WO 2005/014780 PCT/US2004/017231 or higher in the case of individual wells and is 15% or more at the two highest E:T ratios when expanded cultures are assayed.
In situ Measurement of Human IFNy Production as an Indicator of Peptide-specific and Endogenous Recognition [0467] Immulon 2 plates are coated with mouse anti-human IFNy monoclonal antibody (4 g/ml 0.1M NaHCO3, pH8.2) overnight at 4'C. The plates are washed with Ca 2 Mg 2 free PBS/0.05% Tween 20 and blocked with FCS for two hours, after which the CTLs (100 pl/well) and targets (100 pllwell) are added to each well, leaving empty wells for the standards and blanks (which received media only). The target cells, either peptide-pulsed or endogenous targets, are used at a concentration of 1x10 6 cells/ml. The plates are incubated for 48 hours at 37°C with 5% CO2.
[0468] Recombinant human IFN-gamma is added to the standard wells starting at 400 pg or 1200pg/100 microliterlwell and the plate incubated for two hours at 37°C. The plates are washed and 100 pl of biotinylated mouse antihuman IFN-gamma monoclonal antibody (2 microgram/ml in PBS/3%FCS/0.05% Tween 20) are added and incubated for 2 hours at room temperature. After washing again, 100 microliter HRP-streptavidin (1:4000) are added and the plates incubated for one hour at room temperature. The plates are then washed 6x with wash buffer, 100 microliter/well developing solution (TMB 1:1) are added, and the plates allowed to develop for 5-15 minutes. The reaction is stopped with microliter/well 1M H3P04 and read at OD450. A culture is considered positive if it measured at least 50 pg of IFNgamma/well above background and is twice the background level of expression.
CTL Expansion.
[0469] Those cultures that demonstrate specific lytic activity against peptide-pulsed targets and/or tumor targets are expanded over a two week period with anti-CD3. Briefly, 5x10 4 CD8+ cells are added to a T25 flask containing the following: 1x10 6 irradiated (4,200 rad) PBMC (autologous or allogeneic) per ml, 2x10 5 irradiated (8,000 rad) EBVtransformed cells per ml, and OKT3 (anti-CD3) at 30ng per ml in RPMI-1640 containing 10% human AB serum, nonessential amino acids, sodium pyruvate, 25pM 2-mercaptoethanol, L-glutamine and penicillin/streptomycin. Recombinant human IL2 is added 24 hours later at a final concentration of 2001Ulml and every three days thereafter with fresh media at 501U!ml. The cells are split if the cell concentration exceeds lx10 6 /ml and the cultures are assayed between days 13 and at E:T ratios of 30, 10, 3 and 1:1 in the slCr release assay or at 1x10 6 /ml in the in situ IFNy assay using the same targets as before the expansion.
[0470] Cultures are expanded in the absence of anti-CD3* as follows. Those cultures that demonstrate specific lytic activity against peptide and endogenous targets are selected and 5x10 4 CD8+ cells are added to a T25 flask containing the following: 1x10 6 autologous PBMC per ml which have been peptide-pulsed with 10 p.g/ml peptide for two hours at 37°C and irradiated (4,200 rad); 2x10 s irradiated (8,000 rad) EBV-transformed cells per ml RPMI-1640 containing 10%(v/v) human AB serum, non-essential AA, sodium pyruvate, 25mM 2-ME, L-glutamine and gentamicin.
Immunocenicity of A2 supermotif-bearing peptides [0471] A2-supermotif cross-reactive binding peptides are tested in the cellular assay for the ability to induce peptidespecific CTL in normal individuals. In this analysis, a peptide is typically considered to be an epitope if it induces peptidespecific CTLs in at least individuals, and preferably, also recognizes the endogenously expressed peptide.
WO 2005/014780 PCT/US2004/017231 [0472] Immunogenicity can also be confirmed using PBMCs isolated from patients bearing a tumor that expresses PSCA. Briefly, PBMCs are isolated from patients, re-stimulated with peptide-pulsed monocytes and assayed for the ability to recognize peptide-pulsed target cells as well as transfected cells endogenously expressing the antigen.
Evaluation of A*03/A11 immunoaenicity [0473] HLA-A3 supermotif-bearing cross-reactive binding peptides are also evaluated for immunogenicity using methodology analogous for that used to evaluate the immunogenicity of the HLA-A2 supermotif peptides.
Evaluation of B7 immunoqenicity [0474] Immunogenicity screening of the B7-supertype cross-reactive binding peptides identified as set forth herein are confirmed in a manner analogous to the confirmation of A2-and A3-supermotif-bearing peptides.
[0475] Peptides bearing other supermotifs/motifs, HLA-A1, HLA-A24 etc. are also confirmed using similar methodology.
Example 15: Implementation of the Extended Supermotif to Improve the Binding Capacity of Native Epitopes by Creating Analogs [0476] HLA motifs and supermotifs (comprising primary andlor secondary residues) are useful in the identification and preparation of highly cross-reactive native peptides, as demonstrated herein. Moreover, the definition of HLA motifs and supermotifs also allows one to engineer highly cross-reactive epitopes by identifying residues within a native peptide sequence which can be analoged to confer upon the peptide certain characteristics, e.g. greater cross-reactivity within the group of HLA molecules that comprise a supertype, and/or greater binding affinity for some or all of those HLA molecules.
Examples of analoging peptides to exhibit modulated binding affinity are set forth in this example.
Analoqing at Primary Anchor Residues [0477] Peptide engineering strategies are implemented to further increase the cross-reactivity of the epitopes. For example, the main anchors ofA2-supermotif-bearing peptides are altered, for example, to introduce a preferred L, I, V, or M at position 2, and I or V at the C-terminus.
[0478] To analyze the cross-reactivity of the analog peptides, each engineered analog is initially tested for binding to the prototype A2 supertype allele A*0201, then, if A*0201 binding capacity is maintained, for A2-supertype cross-reactivity.
[0479] Alternatively, a peptide is confirmed as binding one or all supertype members and then analoged to modulate binding affinity to any one (or more) of the supertype members to add population coverage.
[0480] The selection of analogs for immunogenicity in a cellular screening analysis is typically further restricted by the capacity of the parent wild type (WT) peptide to bind at least weakly, bind at an IC50 of 5000nM or less, to three of more A2 supertype alleles. The rationale for this requirement is that the WT peptides must be present endogenously in sufficient quantity to be biologically relevant. Analoged peptides have been shown to have increased immunogenicity and cross-reactivity by T cells specific for the parent epitope (see, Parkhurst et J. Immunol. 157:2539, 1996; and Pogue et Proc. Natl. Acad. Sci. USA 92:8166, 1995).
[0481] In the cellular screening of these peptide analogs, it is important to confirm that analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, target cells that endogenously express the epitope.
WO 2005/014780 PCT/US2004/017231 Analoging of HLA-A3 and B7-supermotif-bearing peptides [0482] Analogs of HLA-A3 supermotif-bearing epitopes are generated using strategies similar to those employed in analoging HLA-A2 supermotif-bearing peptides. For example, peptides binding to 3/5 of the A3-supertype molecules are engineered at primary anchor residues to possess a preferred residue S, M, or A) at position 2.
[0483] The analog peptides are then tested for the ability to bind A*03 and A*11 (prototype A3 supertype alleles).
Those peptides that demonstrate 500 nM binding capacity are then confirmed as having A3-supertype cross-reactivity.
[0484] Similarly to the A2- and A3- motif bearing peptides, peptides binding 3 or more B7-supertype alleles can be improved, where possible, to achieve increased cross-reactive binding or greater binding affinity or binding half life. B7 supermotif-bearing peptides are, for example, engineered to possess a preferred residue I, L, or F) at the C-terminal primary anchor position, as demonstrated by Sidney et a. Immunol. 157:3480-3490, 1996).
[0485] Analoging at primary anchor residues of other motif andlor supermotif-bearing epitopes is performed in a like manner.
[0486] The analog peptides are then be confirmed for immunogenicity, typically in a cellular screening assay. Again, it is generally important to demonstrate that analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, targets that endogenously express the epitope.
Analoging at Secondary Anchor Residues [0487] Moreover, HLA supermotifs are of value in engineering highly cross-reactive peptides and/or peptides that bind HLA molecules with increased affinity by identifying particular residues at secondary anchor positions that are associated with such properties. For example, the binding capacity of a B7 supermotif-bearing peptide with an F residue at position 1 is analyzed. The peptide is then analoged to, for example, substitute L for F at position 1. The analoged peptide is evaluated for increased binding affinity, binding half life and/or increased cross-reactivity. Such a procedure identifies analoged peptides with enhanced properties.
[0488] Engineered analogs with sufficiently improved binding capacity or cross-reactivity can also be tested for immunogenicity in HLA-B7-transgenic mice, following for example, IFA immunization or lipopeptide immunization.
Analoged peptides are additionally tested for the ability to stimulate a recall response using PBMC from patients with PSCA-expressing tumors.
Other analoging strategies [0489] Another form of peptide analoging, unrelated to anchor positions, involves the substitution of a cysteine with a-amino butyric acid. Due to its chemical nature, cysteine has the propensity to form disulfide bridges and sufficiently alter the peptide structurally so as to reduce binding capacity. Substitution of a-amino butyric acid for cysteine not only alleviates this problem, but has been shown to improve binding and crossbinding capabilities in some instances (see, e.g., the review by Sette et al., In: Persistent Viral Infections, Eds. R. Ahmed and I. Chen, John Wiley Sons, England, 1999).
[0490] Thus, by the use of single amino acid substitutions, the binding properties and/or cross-reactivity of peptide ligands for HLA supertype molecules can be modulated.
WO 2005/014780 PCT/US2004/017231 Example 16: Identification and confirmation of PSCA-derived sequences with HLA-DR binding motifs [0491] Peptide epitopes bearing an HLA class II supermotif or motif are identified and confirmed as outlined below using methodology similar to that described for HLA Class I peptides.
Selection of HLA-DR-suoermotif-bearing epitopes.
[0492] To identify PSCA-derived, HLA class II HTL epitopes, a PSCA antigen is analyzed for the presence of sequences bearing an HLA-DR-motif or supermotif. Specifically, 15-mer sequences are selected comprising a DRsupermotif, comprising a 9-mer core, and three-residue N- and C-terminal flanking regions (15 amino acids total).
[0493] Protocols for predicting peptide binding to DR molecules have been developed (Southwood et J.
Immunol. 160:3363-3373, 1998). These protocols, specific for individual DR molecules, allow the scoring, and ranking, of 9-mer core regions. Each protocol not only scores peptide sequences for the presence of DR-supermotif primary anchors at position 1 and position 6) within a 9-mer core, but additionally evaluates sequences for the presence of secondary anchors. Using allele-specific selection tables (see, Southwood et al., ibid.), it has been found that these protocols efficiently select peptide sequences with a high probability of binding a particular DR molecule. Additionally, it has been found that performing these protocols in tandem, specifically those for DR1, DR4w4, and DR7, can efficiently select DR cross-reactive peptides.
[0494] The PSCA-derived peptides identified above are tested for their binding capacity for various common HLA- DR molecules. All peptides are initially tested for binding to the DR molecules in the primary panel: DR1, DR4w4, and DR7. Peptides binding at least two of these three DR molecules are then tested for binding to DR2w2 p1, DR2w2 P2, DR6w19, and DR9 molecules in secondary assays. Finally, peptides binding at least two of the four secondary panel DR molecules, and thus cumulatively at least four of seven different DR molecules, are screened for binding to DR4w1 DR5w11, and DR8w2 molecules in tertiary assays. Peptides binding at least seven of the ten DR molecules comprising the primary, secondary, and tertiary screening assays are considered cross-reactive DR binders. PSCA-derived peptides found to bind common HLA-DR alleles are of particular interest.
Selection of DR3 motif peptides [0495] Because HLA-DR3 is an allele that is prevalent in Caucasian, Black, and Hispanic populations, DR3 binding capacity is a relevant criterion in the selection of HTL epitopes. Thus, peptides shown to be candidates may also be assayed for their DR3 binding capacity. However, in view of the binding specificity of the DR3 motif, peptides binding only to DR3 can also be considered as candidates for inclusion in a vaccine formulation.
[0496] To efficiently identify peptides that bind DR3, target PSCA antigens are analyzed for sequences carrying one of the two DR3-speciflc binding motifs reported by Geluk et al. Immunol. 152:5742-5748, 1994). The corresponding peptides are then synthesized and confirmed as having the ability to bind DR3 with an affinity of 1 pM or better, less than 1 Peptides are found that meet this binding criterion and qualify as HLA class II high affinity binders.
[0497] DR3 binding epitopes identified in this manner are included in vaccine compositions with DR supermotifbearing peptide epitopes.
WO 2005/014780 PCT/US2004/017231 [0498] Similarly to the case of HLA class I motif-bearing peptides, the class II motif-bearing peptides are analoged to improve affinity or cross-reactivity. For example, aspartic acid at position 4 of the 9-mer core sequence is an optimal residue for DR3 binding, and substitution for that residue often improves DR 3 binding.
Example 17: Immunogenicity of PSCA-derived HTL epitopes [0499] This example determines immunogenic DR supermotif- and DR3 motif-bearing epitopes among those identified using the methodology set forth herein.
[0500] Immunogenicity of HTL epitopes are confirmed in a manner analogous to the determination of immunogenicity of CTL epitopes, by assessing the ability to stimulate HTL responses and/or by using appropriate transgenic mouse models. Immunogenicity is determined by screening for: in vitro primary induction using normal PBMC or recall responses from patients who have PSCA-expressing tumors.
Example 18: Calculation of Dhenotypic frequencies of HLA-supertypes in various ethnic backgrounds to determine breadth of population coverage [0501] This example illustrates the assessment of the breadth of population coverage of a vaccine composition comprised of multiple epitopes comprising multiple supermotifs and/or motifs.
[0502] In order to analyze population coverage, gene frequencies of HLA alleles are determined. Gene frequencies for each HLA allele are calculated from antigen or allele frequencies utilizing the binomial distribution formulae gf=1- (SQRT(1-at)) (see, Sidney et al., Human Immunol. 45:79-93, 1996). To obtain overall phenotypic frequencies, cumulative gene frequencies are calculated, and the cumulative antigen frequencies derived by the use of the inverse formula [af=1-(1-Cgf) 2 [0503] Where frequency data is not available at the level of DNA typing, correspondence to the serologically defined antigen frequencies is assumed. To obtain total potential supertype population coverage no linkage disequilibrium is assumed, and only alleles confirmed to belong to each of the supertypes are included (minimal estimates). Estimates of total potential coverage achieved by inter-loci combinations are made by adding to the A coverage the proportion of the non-A covered population that could be expected to be covered by the B alleles considered total=A+B*(1-A)).
Confirmed members of the A3-like supertype are A3, All, A31, A*3301, and A*6801. Although the A3-like supertype may also include A34, A66, and A*7401, these alleles were not included in overall frequency calculations. Likewise, confirmed members of the A2-like supertype family are A*0201, A*0202, A*0203, A*0204, A*0205, A*0206, A*0207, A*6802, and A*6901. Finally, the B7-like supertype-confirmed alleles are: B7, B*3501-03, B51, B*5301, B*5401, B*5501-2, B*5601, B*6701, and B*7801 (potentially also B*1401, B*3504-06, B*4201, and B*5602).
[0504] Population coverage achieved by combining the A2-, A3- and B7-supertypes is approximately 86% in five major ethnic groups. Coverage may be extended by including peptides bearing the Al and A24 motifs. On average, Al is present in 12% and A24 in 29% of the population across five different major ethnic groups (Caucasian, North American Black, Chinese, Japanese, and Hispanic). Together, these alleles are represented with an average frequency of 39% in these same ethnic populations. The total coverage across the major ethnicities when Al and A24 are combined with the coverage of the A2-, A3- and B7-supertype alleles is see, Table IV An analogous approach can be used to estimate population coverage achieved with combinations of class II motif-bearing epitopes.
[0505] Immunogenicity studies in humans Bertoni etal., J. Clin. Invest. 100:503, 1997; Doolan etal., Immunity 7:97, 1997; and Threlkeld et J. Immunol. 159:1648, 1997) have shown that highly cross-reactive binding peptides are 103 WO 2005/014780 PCT/US2004/017231 almost always recognized as epitopes. The use of highly cross-reactive binding peptides is an important selection criterion in identifying candidate epitopes for inclusion in a vaccine that is immunogenic in a diverse population.
[0506] With a sufficient number of epitopes (as disclosed herein and from the art), an average population coverage is predicted to be greater than 95% in each of five major ethnic populations. The game theory Monte Carlo simulation analysis, which is known in the art (see Osbome, M.J. and Rubinstein, A. "A course in game theory" MIT Press, 1994), can be used to estimate what percentage of the individuals in a population comprised of the Caucasian, North American Black, Japanese, Chinese, and Hispanic ethnic groups would recognize the vaccine epitopes described herein. A preferred percentage is 90%. A more preferred percentage is Example 19: CTL Recognition Of Endogenously Processed Antigens After Priming [0507] This example confirms that CTL induced by native or analoged peptide epitopes identified and selected as described herein recognize endogenously synthesized, native antigens.
[0508] Effector cells isolated from transgenic mice that are immunized with peptide epitopes, for example HLA-A2 supermotif-bearing epitopes, are re-stimulated in vitro using peptide-coated stimulator cells. Six days later, effector cells are assayed for cytotoxicity and the cell lines that contain peptide-specific cytotoxic activity are further re-stimulated. An additional six days later, these cell lines are tested for cytotoxic activity on 51 Cr labeled Jurkat-A2.1/Kb target cells in the absence or presence of peptide, and also tested on 51 Cr labeled target cells bearing the endogenously synthesized antigen, i.e. cells that are stably transfected with PSCA expression vectors.
[0509] The results demonstrate that CTL lines obtained from animals primed with peptide epitope recognize endogenously synthesized PSCA antigen. The choice of transgenic mouse model to be used for such an analysis depends upon the epitope(s) that are being evaluated. In addition to HLA-A*0201/Kb transgenic mice, several other transgenic mouse models including mice with human All, which may also be used to evaluate A3 epitopes, and B7 alleles have been characterized and others transgenic mice for HLA-A1 and A24) are being developed. HLA-DR1 and HLA-DR3 mouse models have also been developed, which may be used to evaluate HTL epitopes.
Example 20: Activity Of CTL-HTL Conjugated Epitopes In Transgenic Mice [0510] This example illustrates the induction of CTLs and HTLs in transgenic mice, by use of a PSCA-derived CTL and HTL peptide vaccine compositions. The vaccine composition used herein comprise peptides to be administered to a patient with a PSCA-expressing tumor. The peptide composition can comprise multiple CTL and/or HTL epitopes. The epitopes are identified using methodology as described herein. This example also illustrates that enhanced immunogenicity can be achieved by inclusion of one or more HTL epitopes in a CTL vaccine composition; such a peptide composition can comprise an HTL epitope conjugated to a CTL epitope. The CTL epitope can be one that binds to multiple HLA family members at an affinity of 500 nM or less, or analogs of that epitope. The peptides may be lipidated, if desired.
[0511] Immunization procedures: Immunization of transgenic mice is performed as described (Alexander et al., J.
Immunol. 159:4753-4761, 1997). For example, A2/Kh mice, which are transgenic for the human HLA A2.1 allele and are used to confirm the immunogenicity of HLA-A*0201 motif- or HLA-A2 supermotif-bearing epitopes, and are primed subcutaneously (base of the tail) with a 0.1 ml of peptide in Incomplete Freund's Adjuvant, or if the peptide composition is a lipidated CTL/HTL conjugate, in DMSO/saline, or if the peptide composition is a polypeptide, in PBS or Incomplete Freund's Adjuvant. Seven days after priming, splenocytes obtained from these animals are restimulated with syngenic irradiated LPS-activated lymphoblasts coated with peptide.
WO 2005/014780 PCT/US2004/017231 [0512] Cell lines: Target cells for peptide-specific cytotoxicity assays are Jurkat cells transfected with the HLA- A2.1/Kb chimeric gene Vitiello etal., J. Exp. Med. 173:1007, 1991) [0513] In vitro CTL activation: One week after priming, spleen cells (30x10 6 cells/flask) are co-cultured at 37°C with syngeneic, irradiated (3000 rads), peptide coated lymphoblasts (10x10 6 cells/flask) in 10 ml of culture medium/T25 flask.
After six days, effector cells are harvested and assayed for cytotoxic activity.
[0514] Assay for cytotoxic activity: Target cells (1.0 to 1.5x100) are incubated at 37 0 C in the presence of 200 pl of 'Cr. After 60 minutes, dells are washed three times and resuspended in R10 medium. Peptide is added where required at a concentration of 1 pg/ml. For the assay, 10 4 51 Cr-labeled target cells are added to different concentrations of effector cells (final volume of 200 pl) in U-bottom 96-well plates. After a six hour incubation period at 37°C, a 0.1 ml aliquot of supernatant is removed from each well and radioactivity is determined in a Micromedic automatic gamma counter. The percent specific lysis is determined by the formula: percent specific release 100 x (experimental release spontaneous release)/(maximum release spontaneous release). To facilitate comparison between separate CTL assays run under the same conditions, 5 1Cr release data is expressed as lytic units/106 cells. One lytic unit is arbitrarily defined as the number of effector cells required to achieve 30% lysis of 10,000 target cells in a six hour 51 Cr release assay. To obtain specific lytic the lytic units/10 6 obtained in the absence of peptide is subtracted from the lytic units/106 obtained in the presence of peptide. For example, if 30% 51 Cr release is obtained at the effector target ratio of 50:1 5x10 effector cells for 10,000 targets) in the absence of peptide and 5:1 5x10 4 effector cells for 10,000 targets) in the presence of peptide, the specific lytic units would be: [(1/50,000)-(1/500,000)] x 106 18 LU.
[0515] The results are analyzed to assess the magnitude of the CTL responses of animals injected with the immunogenic CTL/HTL conjugate vaccine preparation and are compared to the magnitude of the CTL response achieved using, for example, CTL epitopes as outlined above in the Example entitled "Confirmation of Immunogenicity." Analyses similar to this may be performed to confirm the immunogenicity of peptide conjugates containing multiple CTL epitopes and/or multiple HTL epitopes. In accordance with these procedures, it is found that a CTL response is induced, and concomitantly that an HTL response is induced upon administration of such compositions.
Example 21: Selection of CTL and HTL epitopes for inclusion in a PSCA-specific vaccine.
[0516] This example illustrates a procedure for selecting peptide epitopes for vaccine compositions of the invention.
The peptides in the composition can be in the form of a nucleic acid sequence, either single or one or more sequences minigene) that encodes peptide(s), or can be single and/or polyepitopic peptides.
[0517] The following principles are utilized when selecting a plurality of epitopes for inclusion in a vaccine composition. Each of the following principles is balanced in order to make the selection.
[0518] Epitopes are selected which, upon administration, mimic immune responses that are correlated with PSCA clearance. The number of epitopes used depends on observations of patients who spontaneously clear PSCA. For example, if it has been observed that patients who spontaneously clear PSCA-expressing cells generate an immune response to at least three epitopes from PSCA antigen, then at least three epitopes should be included for HLA class I.
A similar rationale is used to determine HLA class II epitopes.
[0519] Epitopes are often selected that have a binding affinity of an ICso of 500 nM or less for an HLA class I molecule, or for class II, an IC60 of 1000 nM or less; or HLA Class I peptides with high binding scores from the BIMAS web site, at URL bimas.dcrt.nih.govf.
WO 2005/014780 PCT/US2004/017231 [0520] In order to achieve broad coverage of the vaccine through out a diverse population, sufficient supermotif bearing peptides, or a sufficient array of allele-specific motif bearing peptides, are selected to give broad population coverage. In one embodiment, epitopes are selected to provide at least 80% population coverage. A Monte Carlo analysis, a statistical evaluation known in the art, can be employed to assess breadth, or redundancy, of population coverage.
[0521] When creating polyepitopic compositions, or a minigene that encodes same, it is typically desirable to generate the smallest peptide possible that encompasses the epitopes of interest. The principles employed are similar, if not the same, as those employed when selecting a peptide comprising nested epitopes. For example, a protein sequence for the vaccine composition is selected because it has maximal number of epitopes contained within the sequence, ie., it has a high concentration of epitopes. Epitopes may be nested or overlapping frame shifted relative to one another).
For example, with overlapping epitopes, two 9-mer epitopes and one 10-mer epitope can be present in a 10 amino acid peptide. Each epitope can be exposed and bound by an HLA molecule upon administration of such a peptide. A multiepitopic, peptide can be generated synthetically, recombinantly, or via cleavage from the native source. Alternatively, an analog can be made of this native sequence, whereby one or more of the epitopes comprise substitutions that alter the cross-reactivity and/or binding affinity properties of the polyepitopic peptide. Such a vaccine composition is administered for therapeutic or prophylactic purposes. This embodiment provides for the possibility that an as yet undiscovered aspect of immune system processing will apply to the native nested sequence and thereby facilitate the production of therapeutic or prophylactic immune response-inducing vaccine compositions. Additionally such an embodiment provides for the possibility of motif-bearing epitopes for an HLA makeup that is presently unknown. Furthermore, this embodiment (absent the creating of any analogs) directs the immune response to multiple peptide sequences that are actually present in PSCA, thus avoiding the need to evaluate any junctional epitopes. Lastly, the embodiment provides an economy of scale when producing nucleic acid vaccine compositions. Related to this embodiment, computer programs can be derived in accordance with principles in the art, which identify in a target sequence, the greatest number of epitopes per sequence length.
[0522] A vaccine composition comprised of selected peptides, when administered, is safe, efficacious, and elicits an immune response similar in magnitude to an immune response that controls or clears cells that bear or overexpress PSCA.
Example 22: Construction of "Miniqene" Multi-Epitope DNA Plasmids [0523] This example discusses the construction of a minigene expression plasmid. Minigene plasmids may, of course, contain various configurations of B cell, CTL and/or HTL epitopes or epitope analogs as described herein.
[0524] A minigene expression plasmid typically includes multiple CTL and HTL peptide epitopes. In the present example, HLA-A2, -A3, -B7 supermotif-bearing peptide epitopes and HLA-A1 and -A24 motif-bearing peptide epitopes are used in conjunction with DR supermotif-bearing epitopes and/or DR3 epitopes. HLA class I supermotif or motif-bearing peptide epitopes derived PSCA, are selected such that multiple supermotifs/motifs are represented to ensure broad population coverage. Similarly, HLA class II epitopes are selected from PSCA to provide broad population coverage, i.e.
both HLA DR-1-4-7 supermotif-bearing epitopes and HLA DR-3 motif-bearing epitopes are selected for inclusion in the minigene construct. The selected CTL and HTL epitopes are then incorporated into a minigene for expression in an expression vector.
[0525] Such a construct may additionally include sequences that direct the HTL epitopes to the endoplasmic reticulum. For example, the li protein may be fused to one or more HTL epitopes as described in the art, wherein the CLIP 106 WO 2005/014780 PCT/US2004/017231 sequence of the ii protein is removed and replaced with an HLA class II epitope sequence so that HLA class II epitope is directed to the endoplasmic reticulum, where the epitope binds to an HLA class II molecules.
[0526] This example illustrates the methods to be used for construction of a minigene-bearing expression plasmid.
Other expression vectors that may be used for minigene compositions are available and known to those of skill in the art.
[0527] The minigene DNA plasmid of this example contains a consensus Kozak sequence and a consensus murine kappa Ig-light chain signal sequence followed by CTL and/or HTL epitopes selected in accordance with principles disclosed herein. The sequence encodes an open reading frame fused to the Myc and His antibody epitope tag coded for by the pcDNA 3.1 Myc-His vector.
[0528] Overlapping oligonucleotides that can, for example, average about 70 nucleotides in length with 15 nucleotide overlaps, are synthesized and HPLC-purified. The oligonucleotides encode the selected peptide epitopes as well as appropriate linker nucleotides, Kozak sequence, and signal sequence. The final multiepitope minigene is assembled by extending the overlapping oligonucleotides in three sets of reactions using PCR. A Perkin/Elmer 9600 PCR machine is used and a total of 30 cycles are performed using the following conditions: 95°C for 15 sec, annealing temperature below the lowest calculated Tm of each primer pair) for 30 sec, and 72°C for 1 min.
[0529] For example, a minigene is prepared as follows. For a first PCR reaction, 5 pg of each of two oligonucleotides are annealed and extended: In an example using eight oligonucleotides, four pairs of primers, oligonucleotides 1+2, 3+4, 5+6, and 7+8 are combined in 100 pl reactions containing Pfu polymerase buffer (lx= 10 mM KCL, 10 mM (NH4)2S04, 20 mM Tris-chloride, pH 8.75, 2 mM MgSO4, 0.1% Triton X-100, 100 pg/ml BSA), 0.25 mM each dNTP, and 2.5 U of Pfu polymerase. The full-length dimer products are gel-purified, and two reactions containing the product of 1+2 and 3+4, and the product of 5+6 and 7+8 are mixed, annealed, and extended for 10 cycles. Half of the two reactions are then mixed, and 5 cycles of annealing and extension carried out before flanking primers are added to amplify the full length product. The full-length product is gel-purified and cloned into pCR-blunt (Invitrogen) and individual clones are screened by sequencing.
Example 23: The Plasmid Construct and the Degree to Which It Induces Immunogenicity.
[0530] The degree to which a plasmid construct, for example a plasmid constructed in accordance with the previous Example, is able to induce immunogenicity is confirmed in vitro by determining epitope presentation by APC following transduction or transfection of the APC with an epitope-expressing nucleic acid construct. Such a study determines "antigenicity" and allows the use of human APC. The assay determines the ability of the epitope to be presented by the APC in a context that is recognized by a T cell by quantifying the density of epitope-HLA class I complexes on the cell surface. Quantitation can be performed by directly measuring the amount of peptide eluted from the APC (see, Sijts et al., J. Immunol. 156:683-692, 1996; Demotz et al., Nature 342:682-684, 1989); or the number of peptide-HLA class I complexes can be estimated by measuring the amount of lysis or lymphokine release induced by diseased or transfected target cells, and then determining the concentration of peptide necessary to obtain equivalent levels of lysis or lymphokine release (see, Kageyama et J. Immunol. 154:567-576, 1995).
[0531] Alternatively, immunogenicity is confirmed through in vivo injections into mice and subsequent in vitro assessment of CTL and HTL activity, which are analyzed using cytotoxicity and proliferation assays, respectively, as detailed in Alexander et Immunity 1:751-761, 1994.
WO 2005/014780 PCT/US2004/017231 [0532] For example, to confirm the capacity of a DNA minigene construct containing at least one HLA-A2 supermotif peptide to induce CTLs in vivo, HLA-A2.1/Kb transgenic mice, for example, are immunized intramuscularly with 100 Itg of naked cDNA. As a means of comparing the level of CTLs induced by cDNA immunization, a control group of animals is also immunized with an actual peptide composition that comprises multiple epitopes synthesized as a single polypeptide as they would be encoded by the minigene.
[0533] Splenocytes from immunized animals are stimulated twice with each of the respective compositions (peptide epitopes encoded in the minigene or the polyepitopic peptide), then assayed for peptide-specific cytotoxic activity in a 51 Cr release assay. The results indicate the magnitude of the CTL response directed against the A2-restricted epitope, thus indicating the in vivo immunogenicity of the minigene vaccine and polyepitopic vaccine.
[0534] It is, therefore, found that the minigene elicits immune responses directed toward the HLA-A2 supermotif peptide epitopes as does the polyepitopic peptide vaccine. A similar analysis is also performed using other HLA-A3 and HLA-B7 transgenic mouse models to assess CTL induction by HLA-A3 and HLA-B7 motif or supermotif epitopes, whereby it is also found that the minigene elicits appropriate immune responses directed toward the provided epitopes.
[0535] To confirm the capacity of a class II epitope-encoding minigene to induce HTLs in vivo, DR transgenic mice, or for those epitopes that cross react with the appropriate mouse MHC molecule, I-Ab-restricted mice, for example, are immunized intramuscularly with 100 pg of plasmid DNA. As a means of comparing the level of HTLs induced by DNA immunization, a group of control animals is also immunized with an actual peptide composition emulsified in complete Freund's adjuvant. CD4+ T cells, i.e. HTLs, are purified from splenocytes of immunized animals and stimulated with each of the respective compositions (peptides encoded in the minigene). The HTL response is measured using a 3 H-thymidine incorporation proliferation assay, (see, Alexander et al. Immunity 1:751-761, 1994). The results indicate the magnitude of the HTL response, thus demonstrating the in vivo immunogenicity of the minigene.
[0536] DNA minigenes, constructed as described in the previous Example, can also be confirmed as a vaccine in combination with a boosting agent using a prime boost protocol. The boosting agent can consist of recombinant protein Barnett et al., Aids Res. and Human Retroviruses 14, Supplement 3:S299-S309, 1998) or recombinant vaccinia, for example, expressing a minigene or DNA encoding the complete protein of interest (see, Hanke et Vaccine 16:439- 445,1998; Sedegah et al, Proc. Natl. Acad. Sci USA 95:7648-53, 1998; Hanke and McMichael, Immunol. Letters 66:177- 181, 1999; and Robinson et al, Nature Med. 5:526-34, 1999).
[0537] For example, the efficacy of the DNA minigene used in a prime boost protocol is initially evaluated in transgenic mice. In this example, A2.1/Kb transgenic mice are immunized IM with 100 gg of a DNA minigene encoding the immunogenic peptides including at least one HLA-A2 supermotif-bearing peptide. After an incubation period (ranging from 3-9 weeks), the mice are boosted IP with 107 pfulmouse of a recombinant vaccinia virus expressing the same sequence encoded by the DNA minigene. Control mice are immunized with 100 p.g of DNA or recombinant vaccinia without the minigene sequence, or with DNA encoding the minigene, but without the vaccinia boost. After an additional incubation period of two weeks, splenocytes from the mice are immediately assayed for peptide-specific activity in an ELISPOT assay.
Additionally, splenocytes are stimulated in vitro with the A2-restricted peptide epitopes encoded in the minigene and recombinant vaccinia, then assayed for peptide-specific activity in an alpha, beta and/or gamma IFN ELISA.
[0538] It is found that the minigene utilized in a prime-boost protocol elicits greater immune responses toward the HLA-A2 supermotif peptides than with DNA alone. Such an analysis can also be performed using HLA-A11 or HLA-B7 transgenic mouse models to assess CTL induction by HLA-A3 or HLA-B7 motif or supermotif epitopes. The use of prime WO 2005/014780 PCT/US2004/017231 boost protocols in humans is described below in the Example entitled "Induction of CTL Responses Using a Prime Boost Protocol." Example 24: Peptide Compositions for Prophylactic Uses [0539] Vaccine compositions of the present invention can be used to prevent PSCA expression in persons who are at risk for tumors that bear this antigen. For example, a polyepitopic peptide epitope composition (or a nucleic acid comprising the same) containing multiple CTL and HTL epitopes such as those selected in the above Examples, which are also selected to target greater than 80% of the population, is administered to individuals at risk for a PSCA-associated tumor.
[0540] For example, a peptide-based composition is provided as a single polypeptide that encompasses multiple epitopes. The vaccine is typically administered in a physiological solution that comprises an adjuvant, such as Incomplete Freunds Adjuvant. The dose of peptide for the initial immunization is from about 1 to about 50,000 p.g, generally 100-5,000 [ig, for a 70 kg patient. The initial administration of vaccine is followed by booster dosages at 4 weeks followed by evaluation of the magnitude of the immune response in the patient, by techniques that determine the presence of epitopespecific CTL populations in a PBMC sample. Additional booster doses are administered as required. The composition is found to be both safe and efficacious as a prophylaxis against PSCA-associated disease.
[0541] Alternatively, a composition typically comprising transfecting agents is used for the administration of a nucleic acid-based vaccine in accordance with methodologies known in the art and disclosed herein.
Example 25: Polvepitopic Vaccine Compositions Derived from Native PSCA Sequences [0542] A native PSCA polyprotein sequence is analyzed, preferably using computer algorithms defined for each class I and/or class II supermotif or motif, to identify "relatively short" regions of the polyprotein that comprise multiple epitopes. The "relatively short" regions are preferably less in length than an entire native antigen. This relatively short sequence that contains multiple distinct or overlapping, "nested" epitopes can be used to generate a minigene construct.
The construct is engineered to express the peptide, which corresponds to the native protein sequence. The "relatively short" peptide is generally less than 250 amino acids in length, often less than 100 amino acids in length, preferably less than 75 amino acids in length, and more preferably less than 50 amino acids in length. The protein sequence of the vaccine composition is selected because it has maximal number of epitopes contained within the sequence, it has a high concentration of epitopes. As noted herein, epitope motifs may be nested or overlapping frame shifted relative to one another). For example, with overlapping epitopes, two 9-mer epitopes and one 10-mer epitope can be present in a amino acid peptide. Such a vaccine composition is administered for therapeutic or prophylactic purposes.
[0543] The vaccine composition will include, for example, multiple CTL epitopes from PSCA antigen and at least one HTL epitope. This polyepitopic native sequence is administered either as a peptide or as a nucleic acid sequence which encodes the peptide. Alternatively, an analog can be made of this native sequence, whereby one or more of the epitopes comprise substitutions that alter the cross-reactivity and/or binding affinity properties of the polyepitopic peptide.
[0544] The embodiment of this example provides for the possibility that an as yet undiscovered aspect of immune system processing will apply to the native nested sequence and thereby facilitate the production of therapeutic or prophylactic immune response-inducing vaccine compositions. Additionally, such an embodiment provides for the WO 2005/014780 PCT/US2004/017231 possibility of motif-bearing epitopes for an HLA makeup(s) that is presently unknown. Furthermore, this embodiment (excluding an analoged embodiment) directs the immune response to multiple peptide sequences that are actually present in native PSCA, thus avoiding the need to evaluate any junctional epitopes. Lastly, the embodiment provides an economy of scale when producing peptide or nucleic acid vaccine compositions.
[0545] Related to this embodiment, computer programs are available in the art which can be used to identify in a target sequence, the greatest number of epitopes per sequence length.
Example 26: Polvepitopic Vaccine Compositions from Multiple Anticens [0546] The PSCA peptide epitopes of the present invention are used in conjunction with epitopes from other target tumor-associated antigens, to create a vaccine composition that is useful for the prevention or treatment of cancer that expresses PSCA and such other antigens. For example, a vaccine composition can be provided as a single polypeptide that incorporates multiple epitopes from PSCA as well as tumor-associated antigens that are often expressed with a target cancer associated with PSCA expression, or can be administered as a composition comprising a cocktail of one or more discrete epitopes. Alteratively, the vaccine can be administered as a minigene construct or as dendritic cells which have been loaded with the peptide epitopes in vitro.
Example 27: Use of peptides to evaluate an immune response [0547] Peptides of the invention may be used to analyze an immune response for the presence of specific antibodies, CTL or HTL directed to PSCA. Such an analysis can be performed in a manner described by Ogg et a., Science 279:2103-2106, 1998. In this Example, peptides in accordance with the invention are used as a reagent for diagnostic or prognostic purposes, not as an immunogen.
[0548] In this example highly sensitive human leukocyte antigen tetrameric complexes ("tetramers") are used for a cross-sectional analysis of, for example, PSCA HLA-A*0201-specific CTL frequencies from HLA A*0201-positive individuals at different stages of disease or following immunization comprising a PSCA peptide containing an A*0201 motif. Tetrameric complexes are synthesized as described (Musey et al., N. Engl. J. Med. 337:1267, 1997). Briefly, purified HLA heavy chain (A*0201 in this example) and j2-microglobulin are synthesized by means of a prokaryotic expression system. The heavy chain is modified by deletion of the transmembrane-cytosolic tail and COOH-terminal addition of a sequence containing a BirA enzymatic biotinylation site. The heavy chain, p2-microglobulin, and peptide are refolded by dilution. The refolded product is isolated by fast protein liquid chromatography and then biotinylated by BirA in the presence of biotin (Sigma, St. Louis, Missouri), adenosine 5' triphosphate and magnesium. Streptavidin-phycoerythrin conjugate is added in a 1:4 molar ratio, and the tetrameric product is concentrated to 1 mg/ml. The resulting product is referred to as tetramerphycoerythrin.
[0549] For the analysis of patient blood samples, approximately one million PBMCs are centrifuged at 300g for minutes and resuspended in 50 pl of cold phosphate-buffered saline. Tri-color analysis is performed with the tetramerphycoerythrin, along with anti-CD8-Tricolor, and anti-CD38. The PBMCs are incubated with tetramer and antibodies on ice for 30 to 60 min and then washed twice before formaldehyde fixation. Gates are applied to contain >99.98% of control samples. Controls for the tetramers include both A*0201-negative individuals and A*0201-positive non-diseased donors.
The percentage of cells stained with the tetramer is then determined by flow cytometry. The results indicate the number of cells in the PBMC sample that contain epitope-restricted CTLs, thereby readily indicating the extent of immune response to WO 2005/014780 PCT/US2004/017231 the PSCA epitope, and thus the status of exposure to PSCA, or exposure to a vaccine that elicits a protective or therapeutic response.
Example 28: Use of Peptide Epitopes to Evaluate Recall Responses [0550] The peptide epitopes of the invention are used as reagents to evaluate T cell responses, such as acute or recall responses, in patients. Such an analysis may be performed on patients who have recovered from PSCA-associated disease or who have been vaccinated with a PSCA vaccine.
[0551] For example, the class I restricted CTL response of persons who have been vaccinated may be analyzed.
The vaccine may be any PSCA vaccine. PBMC are collected from vaccinated individuals and HLA typed. Appropriate peptide epitopes of the invention that, optimally, bear supermotifs to provide cross-reactivity with multiple HLA supertype family members, are then used for analysis of samples derived from individuals who bear that HLA type.
[0552] PBMC from vaccinated individuals are separated on Ficoll-Histopaque density gradients (Sigma Chemical Co., St. Louis, MO), washed three times in HBSS (GIBCO Laboratories), resuspended in RPMI-1640 (GIBCO Laboratories) supplemented with L-glutamine (2mM), penicillin (50U/ml), streptomycin (50 1pg/ml), and Hepes (10mM) containing heat-inactivated human AB serum (complete RPMI) and plated using microculture formats. A synthetic peptide comprising an epitope of the invention is added at 10 1 ig/ml to each well and HBV core 128-140 epitope is added at 1 tglml to each well as a source of T cell help during the first week of stimulation.
[0553] In the microculture format, 4 x 105 PBMC are stimulated with peptide in 8 replicate cultures in 96-well round bottom plate in 100 illwell of complete RPMI. On days 3 and 10, 100 pl of complete RPMI and 20 UIml final concentration of rlL-2 are added to each well. On day 7 the cultures are transferred into a 96-well flat-bottom plate and restimulated with peptide, rlL-2 and 105 irradiated (3,000 rad) autologous feeder cells. The cultures are tested for cytotoxic activity on day 14. A positive CTL response requires two or more of the eight replicate cultures to display greater than 10% specific 51 Cr release, based on comparison with non-diseased control subjects as previously described (Rehermann, et al., Nature Med.
2:1104,1108, 1996; Rehermann et al., J. Clin. Invest. 97:1655-1665, 1996; and Rehermann etal. J. Clin. Invest. 98:1432- 1440,1996).
[0554] Target cell lines are autologous and allogeneic EBV-transformed B-LCL that are either purchased from the American Society for Histocompatibility and Immunogenetics (ASHI, Boston, MA) or established from the pool of patients as described (Guilhot, et al. J. Virol. 66:2670-2678, 1992).
[0555] Cytotoxlcity assays are performed in the following manner. Target cells consist of either allogeneic HLAmatched or autologous EBV-transformed B lymphoblastoid cell line that are incubated overnight with the synthetic peptide epitope of the invention at 10 IM, and labeled with 100 pCi of 51Cr (Amersham Corp., Arlington Heights, IL) for 1 hour after which they are washed four times with HBSS.
[0556] Cytolytic activity is determined in a standard 4-h, split well 51 Cr release assay using U-bottomed 96 well plates containing 3,000 targets/well. Stimulated PBMC are tested at effector/target ratios of 20-50:1 on day 14.
Percent cytotoxicity is determined from the formula: 100 x [(experimental release-spontaneous release)/maximum releasespontaneous release)]. Maximum release is determined by lysis of targets by detergent Triton X-100; Sigma Chemical Co., St. Louis, MO). Spontaneous release is <25% of maximum release for all experiments.
[0557] The results of such an analysis indicate the extent to which HLA-restricted CTL populations have been stimulated by previous exposure to PSCA or a PSCA vaccine.
WO 2005/014780 PCT/US2004/017231 [0558] Similarly, Class II restricted HTL responses may also be analyzed. Purified PBMC are cultured in a 96-well flat bottom plate at a density of 1.5x105 cells/well and are stimulated with 10 p.g/ml synthetic peptide of the invention, whole PSCA antigen, or PHA. Cells are routinely plated in replicates of 4-6 wells for each condition. After seven days of culture, the medium is removed and replaced with fresh medium containing 10U/ml IL-2. Two days later, 1 pCi 3 H-thymidine is added to each well and incubation is continued for an additional 18 hours. Cellular DNA is then harvested on glass fiber mats and analyzed for 3 H-thymidine incorporation. Antigen-specific T cell proliferation is calculated as the ratio of 3
H-
thymidine incorporation in the presence of antigen divided by the 3 H-thymidine incorporation in the absence of antigen.
Example 29: Induction Of Specific CTL Response In Humans [0559] A human clinical trial for an immunogenic composition comprising CTL and HTL epitopes of the invention is set up as an IND Phase I, dose escalation study and carried out as a randomized, double-blind, placebo-controlled trial.
Such a trial is designed, for example, as follows: [0560] A total of about 27 individuals are enrolled and divided into 3 groups: [0561] Group I: 3 subjects are injected with placebo and 6 subjects are injected with 5 p.g of peptide composition; [0562] Group 11: 3 subjects are injected with placebo and 6 subjects are injected with 50 pg peptide composition; [0563] Group III: 3 subjects are injected with placebo and 6 subjects are injected with 500 p.g of peptide composition.
[0564] After 4 weeks following the first injection, all subjects receive a booster inoculation at the same dosage.
[0565] The endpoints measured in this study relate to the safety and tolerability of the peptide composition as well as its immunogenicity. Cellular immune responses to the peptide composition are an index of the intrinsic activity of this the peptide composition, and can therefore be viewed as a measure of biological efficacy. The following summarize the clinical and laboratory data that relate to safety and efficacy endpoints.
[0566] Safety: The incidence of adverse events is monitored in the placebo and drug treatment group and assessed in terms of degree and reversibility.
[0567] Evaluation of Vaccine Efficacy: For evaluation of vaccine efficacy, subjects are bled before and after injecton. Peripheral blood mononuclear cells are isolated from fresh heparinized blood by Ficoll-Hypaque density gradient centrifugation, aliquoted in freezing media and stored frozen. Samples are assayed for CTL and HTL activity.
[0568] The vaccine is found to be both safe and efficacious.
Example 30: Phase II Trials In Patients Expressing PSCA [0569] Phase II trials are performed to study the effect of administering the CTL-HTL peptide compositions to patients having cancer that expresses PSCA. The main objectives of the trial are to determine an effective dose and regimen for inducing CTLs in cancer patients that express PSCA, to establish the safety of inducing a CTL and HTL response in these patients, and to see to what extent activation of CTLs improves the clinical picture of these patients, as manifested, by the reduction and/or shrinking of lesions. Such a study is designed, for example, as follows: [0570] The studies are performed in multiple centers. The trial design is an open-label, uncontrolled, dose escalation protocol wherein the peptide composition is administered as a single dose followed six weeks later by a single booster shot of the same dose. The dosages are 50, 500 and 5,000 micrograms per injection. Drug-associated adverse effects (severity and reversibility) are recorded.
WO 2005/014780 PCT/US2004/017231 [0571] There are three patient groupings. The first group is injected with 50 micrograms of the peptide composition and the second and third groups with 500 and 5,000 micrograms of peptide composition, respectively. The patients within each group range in age from 21-65 and represent diverse ethnic backgrounds. All of them have a tumor that expresses
PSCA.
[0572] Clinical manifestations or antigen-specific T-cell responses are monitored to assess the effects of administering the peptide compositions. The vaccine composition is found to be both safe and efficacious in the treatment of PSCA-associated disease.
Example 31: Induction of CTL Responses Using a Prime Boost Protocol [0573] A prime boost protocol similar in its underlying principle to that used to confirm the efficacy of a DNA vaccine in transgenic mice, such as described above in the Example entitled "The Plasmid Construct and the Degree to Which It Induces Immunogenicity," can also be used for the administration of the vaccine to humans. Such a vaccine regimen can include an initial administration of, for example, naked DNA followed by a boost using recombinant virus encoding the vaccine, or recombinant protein/polypeptide or a peptide mixture administered in an adjuvant.
[0574] For example, the initial immunization may be performed using an expression vector, such as that constructed in the Example entitled "Construction of "Minigene" Multi-Epitope DNA Plasmids" in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites. The nucleic acid (0.1 to 1000 jig) can also be administered using a gene gun. Following an incubation period of 3-4 weeks, a booster dose is then administered. The booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5x109 pfu. An alternative recombinant virus, such as an MVA, canarypox, adenovirus, or adeno-associated virus, can also be used for the booster, or the polyepitopic protein or a mixture of the peptides can be administered. For evaluation of vaccine efficacy, patient blood samples are obtained before immunization as well as at intervals following administration of the initial vaccine and booster doses of the vaccine. Peripheral blood mononuclear cells are isolated from fresh heparinized blood by Ficoll-Hypaque density gradient centrifugation, aliquoted in freezing media and stored frozen. Samples are assayed for CTL and HTL activity.
[0575] Analysis of the results indicates that a magnitude of response sufficient to achieve a therapeutic or protective immunity against PSCA is generated.
Example 32: Administration of Vaccine Compositions Using Dendritic Cells (DC) [0576] Vaccines comprising peptide epitopes of the invention can be administered using APCs, or "professional" APCs such as DC. In this example, peptide-pulsed DC are administered to a patient to stimulate a CTL response in vivo.
In this method, dendritic cells are isolated, expanded, and pulsed with a vaccine comprising peptide CTL and HTL epitopes of the invention. The dendritic cells are infused back into the patient to elicit CTL and HTL responses in vivo. The induced CTL and HTL then destroy or facilitate destruction, respectively, of the target cells that bear the PSCA protein from which the epitopes in the vaccine are derived.
[0577] For example, a cocktail of epitope-comprising peptides is administered ex vivo to PBMC, or isolated DC therefrom. A pharmaceutical to facilitate harvesting of DC can be used, such as ProgenipoietinT (Monsanto, St. Louis, MO) or GM-CSF/IL-4. After pulsing the DC with peptides, and prior to reinfusion into patients, the DC are washed to remove unbound peptides.
WO 2005/014780 PCT/US2004/017231 [0578] As appreciated clinically, and readily determined by one of skill based on clinical outcomes, the number of DC reinfused into the patient can vary (see, Nature Med. 4:328, 1998; Nature Med. 2:52, 1996 and Prostate 32:272, 1997). Although 2-50 x 106 DC per patient are typically administered, larger number of DC, such as 107 or 10 B can also be provided. Such cell populations typically contain between 50-90% DC.
[0579] In some embodiments, peptide-loaded PBMC are injected into patients without purification of the DC. For example, PBMC generated after treatment with an agent such as PrcgenipoetinM are Injected into patients without purification of the DC. The total number of PBMC that are administered often ranges from 108 to 1010. Generally, the cell doses injected into patients is based on the percentage of DC in the blood of each patient, as determined, for example, by immunofluorescence analysis with specific anti-DC antibodies. Thus, for example, if ProgenipoietinT mobilizes 2% DC in the peripheral blood of a given patient, and that patient is to receive 5 x 106 DC, then the patient will be injected with a total of 2.5 x 108 peptide-loaded PBMC. The percent DC mobilized by an agent such as Progenipoietin T M is typically estimated to be between 2-10%, but can vary as appreciated by one of skill in the art.
Ex vivo activation of CTL/HTL responses [0580] Alternatively, ex vivo CTL or HTL responses to PSCA antigens can be induced by incubating, in tissue culture, the patient's, or genetically compatible, CTL or HTL precursor cells together with a source of APC, such as DC, and immunogenic peptides. After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cells, tumor cells.
Example 33: An Alternative Method of Identifving and Confirminq Motif-Bearing Peptides [0581] Another method of identifying and confirming motif-bearing peptides is to elute them from cells bearing defined MHC molecules. For example, EBV transformed B cell lines used for tissue typing have been extensively characterized to determine which HLA molecules they express. In certain cases these cells express only a single type of HLA molecule. These cells can be transfected with nucleic acids that express the antigen of interest, e.g. PSCA. Peptides produced by endogenous antigen processing of peptides produced as a result of transfection will then bind to HLA molecules within the cell and be transported and displayed on the cell's surface. Peptides are then eluted from the HLA molecules by exposure to mild acid conditions and their amino acid sequence determined, by mass spectral analysis Kubo et J. Immunol. 152:3913,1994). Because the majority of peptides that bind a particular HLA molecule are motif-bearing, this is an alternative modality for obtaining the motif-bearing peptides correlated with the particular HLA molecule expressed on the cell.
[0582] Alternatively, cell lines that do not express endogenous HLA molecules can be transfected with an expression construct encoding a single HLA allele. These cells can then be used as described, they can then be transfected with nucleic acids that encode PSCA to isolate peptides corresponding to PSCA that have been presented on the cell surface.
Peptides obtained from such an analysis will bear motif(s) that correspond to binding to the single HLA allele that is expressed in the cell.
[0583] As appreciated by one in the art, one can perform a similar analysis on a cell bearing more than one HLA allele and subsequently determine peptides specific for each HLA allele expressed. Moreover, one of skill would also WO 2005/014780 PCT/US2004/017231 recognize that means other than transfection, such as loading with a protein antigen, can be used to provide a source of antigen to the cell.
Example 34: Complementary Polynucleotides [0584] Sequences complementary to the PSCA-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring PSCA. Although use of ollgonucleotides comprising from about 15 to base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments.
Appropriate oligonucleotides are designed using, OLIGO 4.06 software (National Biosciences) and the coding sequence of PSCA. To inhibit transcription, a complementary oligonucleotide is designed from the most unique sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to a PSCA-encoding transcript.
Example 35: Purification of Naturally-occurring or Recombinant PSCA Using PSCA- Specific Antibodies [0585] Naturally occurring or recombinant PSCA is substantially purified by immunoaffinity chromatography using antibodies specific for PSCA. An immunoaffinity column is constructed by covalently coupling anti-PSCA antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Pharmacia Biotech). After the coupling, the resin is blocked and washed according to the manufacturer's instructions.
[0586] Media containing PSCA are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of PSCA high ionic strength buffers in the presence of detergent).
The column is eluted under conditions that disrupt antibody/PSCA binding a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and GCR.P is collected.
Example 36: Identification of'Molecules Which Interact with PSCA [0587] PSCA, or biologically active fragments thereof, are labeled with 1211 Bolton-Hunter reagent. (See, e.g., Bolton et al. (1973) Biochem. J. 133:529.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled PSCA, washed, and any wells with labeled PSCA complex are assayed. Data obtained using different concentrations of PSCA are used to calculate values for the number, affinity, and association of PSCA with the candidate molecules.
Example 37: In Vivo Assay for PSCA v.4 Tumor Growth Promotion [0588] The effect of the PSCA v.4 protein on tumor cell growth is evaluated in vivo by evaluating tumor development and growth of cells expressing or lacking PSCA v.4. For example, SCID mice are injected subcutaneously on each flank with 1 x 10 5 of either 3T3, prostate PC3 cells), bladder UM-UC3 cells) or pancreas PANC1 cells) cancer cell lines containing tkNeo empty vector or PSCA v.4. At least two strategies may be used: Constitutive PSCA v.4 expression under regulation of a promoter such as a constitutive promoter obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 July 1989), adenovirus (such as Adenovirus bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), or from heterologous mammalian promoters, the actin promoter or an immunoglobulin promoter, provided such promoters are WO 2005/014780 PCT/US2004/017231 compatible with the host cell systems, and Regulated expression under control of an inducible vector system, such as ecdysone, tetracycline, etc., provided such promoters are compatible with the host cell systems. Tumor volume is then monitored by caliper measurement at the appearance of palpable tumors and followed over time to determine if PSCA v.4expressing cells grow at a faster rate and whether tumors produced by PSCA v.4-expressing cells demonstrate characteristics of altered aggressiveness enhanced metastasis, vascularization, reduced responsiveness to chemotherapeutic drugs).
[0589] Additionally, mice can be implanted with 1 x 105 of the same cells orthotopically to determine if PSCA v.4 has an effect on local growth in the pancreas, and whether PSCA v.4 affects the ability of the cells to metastasize, specifically to lymph nodes, and bone (Miki Tet al, Oncol Res. 2001;12:209; Fu Xetal, Int. J Cancer. 1991, 49:938). The effect of PSCA v.4 on bone tumor formation and growth may be assessed by injecting tumor cells intratibially.
[0590] The assay is also useful to determine the PSCA v.4 inhibitory effect of candidate therapeutic compositions, such as for example, PSCA v.4 intrabodies, PSCA v.4 antisense molecules and ribozymes.
Example 38: PSCA v.4 Monoclonal Antibody-mediated Inhibition of Tumors In Vivo [0591] The significant expression of PSCA v.4 in cancer tissues, together with its restrictive expression in normal tissues makes PSCA v.4 a good target for antibody therapy. Similarly, PSCA v.4 is a target for T cell-based immunotherapy. Thus, the therapeutic efficacy of anti-PSCA v.4 mAbs in human cancer xenograft mouse models, including prostate, bladder, and pancreas PANC1 cells) and other -PSCA v.4 cancers listed in table 1, is evaluated by using recombinant cell lines such as PC3-PSCAv.4, UM-UC3-PSCA v.4, PANC1-PSCA v.4, and 3T3-PSCA v.4 (see, e.g., Kaighn, et al., Invest Urol, 1979.17(1): 16-23), as well as human xenograft models (Saffran et al PNAS 1999, 10:1073-1078).
[0592] Antibody efficacy on tumor growth and metastasis formation is studied, in a mouse orthotopic ovary, pancreas, or blood cancer xenograft models. The antibodies can be unconjugated, as discussed in this Example, or can be conjugated to a therapeutic modality, as appreciated in the art. Anti-PSCA v.4 mAbs inhibit formation of tumors in mouse xenografts. Anti-PSCA v.4 mAbs also retard the growth of established orthotopic tumors and prolonged survival of tumorbearing mice. These results indicate the utility of anti-PSCA v.4 mAbs in the treatment of local and advanced stages several solid tumors. (See, Saffran, et al., PNAS 10:1073-1078 or world wide web URL pnas.org/cgildoi/10.1073/pnas.051624698).
[0593] Administration of the anti-PSCA v.4 mAbs led to retardation of established orthotopic tumor growth and inhibition of metastasis to distant sites, resulting in a significant prolongation in the survival of tumor-bearing mice. These studies indicate that PSCA v.4 as an attractive target for immunotherapy and demonstrate the therapeutic potential of anti- PSCA v.4 mAbs for the treatment of local and metastatic cancer. This example indicates that unconjugated PSCA v.4 monoclonal antibodies are effective to inhibit the growth of human pancreatic, ovarian, and lymphomas tumor xenografts grown in SCID mice; accordingly a combination of such efficacious monoclonal antibodies is also effective.
WO 2005/014780 PCT/US2004/017231 Tumor inhibition using multiple unconiugated PSCA v.4 mAbs Materials and Methods PSCA v.4 Monoclonal Antibodies: [0594] Monoclonal antibodies are raised against PSCA v.4 as described in the Example entitled "Generation of PSCA v.4 Monoclonal Antibodies (mAbs)." The antibodies are characterized by ELISA, Western blot, FACS, and immunoprecipitation for their capacity to bind PSCA v.4. Epitope mapping data for the anti-PSCA v.4 mAbs, as determined by ELISA and Western analysis, recognize epitopes on the PSCA v.4 protein. Immunohistochemical analysis of cancer tissues and cells with these antibodies is performed.
[0595] The monoclonal antibodies are purified from ascites or hybridoma tissue culture supernatants by Protein-G Sepharose chromatography, dialyzed against PBS, filter sterilized, and stored at -20oC. Protein determinations are performed by a Bradford assay (Bio-Rad, Hercules, CA). A therapeutic monoclonal antibody or a cocktail comprising a mixture of individual monoclonal antibodies is prepared and used for the treatment of mice receiving subcutaneous or orthotopic injections of PC3, UM-UC3, CaKi, and A427 tumor xenografts.
Cell Lines and Xenoarafts [0596] The LAPC-9 xenograft, which expresses a wild-type androgen receptor and produces prostate-specific antigen (PSA), is passaged in 6- to 8-week-old male ICR-severe combined immunodeficient (SCID) mice (Taconic Farms) by s.c. trocar implant (Craft, et al., 1999, Cancer Res. 59:5030-5036). The AGS-K3 and AGS-K6 kidney xenografts are also passaged by subcutaneous implants in 6- to 8- week old SCID mice. Single-cell suspensions of tumor cells are prepared as described in Craft, et al.
[0597] The cancer cell lines PC3, UM-UC3 and PANC1 cell lines, as well as the fibroblast line NIH 3T3 (American Type Culture Collection). The prostate carcinoma cell line PC3 is maintained in RPMI supplemented with L-glutamine and FBS, and the bladder and pancreas carcinoma lines, UM-UC3 and PANC1 respectively, are maintained in DMEM supplemented with L-glutamine and 10% FBS. PC3-PSCA v.4, UM-UC3-PSCA v.4, PANC1-PSCA v.4 and 3T3-PSCA v.4 cell populations are generated by retroviral gene transfer as described in Hubert, et al., Proc Natl. Acad. Sc U S A, 1999. 96(25): 14523.
Xenograft Mouse Models.
[0598] Subcutaneous tumors are generated by injection of 2 x 10 6 cancer cells mixed at a 1:1 dilution with Matrigel (Collaborative Research) in the right flank of male SCID mice. To test antibody efficacy on tumor formation, i.e.
antibody injections are started on the same day as tumor-cell injections. As a control, mice are injected with either purified mouse IgG (ICN) or PBS; or a purified monoclonal antibody that recognizes an irrelevant antigen not expressed in human cells. In preliminary studies, no difference is found between mouse IgG or PBS on tumor growth. Tumor sizes are determined by caliper measurements, and the tumor volume is calculated as length x width x height. Mice with Subcutaneous tumors greater than 1.5 cm in diameter are sacrificed.
[0599] Orthotopic injections are performed under anesthesia by using ketamine/xylazine.. For prostate orthotopic studies, an incision is made through the abdominal muscles to expose the bladder and seminal vesicles, which then are delivered through the incision to expose the dorsal prostate. LAPC-9 cells (5 x 105) mixed with Matrigel are injected into WO 2005/014780 PCT/US2004/017231 each dorsal lobe in a 10 pl volume. To monitor tumor growth, mice are bled on a weekly basis for determination of PSA levels. For pancreas orthotoptic model, an incision is made through the abdominal muscles to expose the mammary tissues and a single cell suspension of pancreas cancer cells is injected into the mammary pad. For the bladder orthotopic model, AGS-B1 bladder cancer tissue is adhered onto the bladder wall. Following tumor implantation, the mice are segregated into groups for the appropriate treatments, with anti-PSCA v.4 or control mAbs being injected i.p. To monitor tumor growth, mice are palpated and blood is collected on a weekly basis to measure hCG levels.
Anti-PSCA v.4 mAbs Inhibit Growth of PSCA v.4-Expressing Xenocraft-Cancer Tumors [0600] The effect of anti-PSCA v.4 mAbs on tumor formation is tested by using cell line PC3, UM-UC3, PANC1 and 3T3) and patient-derived tumor orthotopic models. As compared with the s.c. tumor model, the orthotopic model, which requires injection of tumor cells directly in the mouse organ results in a local tumor growth, development of metastasis in distal sites, deterioration of mouse health, and subsequent death (Saffran, et al., PNAS supra). The features make the orthotopic model more representative of human disease progression and allowed us to follow the therapeutic effect of mAbs on clinically relevant end points.
[0601] A major advantage of the orthotopic cancer models is the ability to study the development of metastases.
Formation of metastasis in mice bearing established orthotopic tumors is studies by IHC analysis on lung sections using an antibody against a tumor-specific cell-surface protein such as anti-CK20 for prostate cancer (Lin S et al, Cancer Detect Prev. 2001;25:202).
[0602] Another advantage of xenograft cancer models is the ability to study neovascularization and angicgenesis.
Tumor growth is partly dependent on new blood vessel development. Although the capillary system and developing blood network is of host origin, the initiation and architecture of the neovasculature is regulated by the xenograft tumor (Davidoff AM et al, Clin Cancer Res. 2001;7:2870; Solesvik O et al,, Eur. J Cancer Clin Oncol. 1984, 20:1295). The effect of antibody and small molecule on neovascularization is studied in accordance with procedures known in the art, such as by IHC analysis of tumor tissues and their surrounding microenvironment.
[0603] Mice bearing established orthotopic tumors are administered 1000pg injections of either anti-PSCA v.4 mAb or PBS over a 4-week period. Mice in both groups are allowed to establish a high tumor burden, to ensure a high frequency of metastasis formation in mouse lungs. Mice then are killed and their bladders, livers, bone and lungs are analyzed for the presence of tumor cells by IHC analysis. These studies demonstrate a broad anti-tumor efficacy of anti- PSCA v.4 antibodies on initiation and progression of prostate cancer in xenograft mouse models. Anti-PSCA v.4 antibodies inhibit tumor formation of tumors as well as retarding the growth of already established tumors and prolong the survival of treated mice. Moreover, anti-PSCA v.4 mAbs demonstrate a dramatic inhibitory effect on the spread of local prostate tumor to distal sites, even in the presence of a large tumor burden. Thus, anti-PSCA v.4 mAbs are efficacious on major clinically relevant end points (tumor growth), prolongation of survival, and health.
Example 39: Therapeutic and Diagnostic use of Anti-PSCA Antibodies in Humans.
[0604] Anti-PSCA monoclonal antibodies are safely and effectively used for diagnostic, prophylactic, prognostic and/or therapeutic purposes in humans. Western blot and immunohistochemical analysis of cancer tissues and cancer xenografts with anti-PSCA mAb show strong extensive staining in carcinoma but significantly lower or undetectable levels in normal tissues. Detection of PSCA in carcinoma and in metastatic disease demonstrates the usefulness of the mAb as a 118 WO 2005/014780 PCT/US2004/017231 diagnostic and/or prognostic indicator. Anti-PSCA antibodies are therefore used in diagnostic applications such as immunohistochemistry of kidney biopsy specimens to detect cancer from suspect patients.
[0605] As determined by flow cytometry, anti-PSCA mAb specifically binds to carcinoma cells. Thus, anti-PSCA antibodies are used in diagnostic whole body imaging applications, such as radioimmunoscintigraphy and radioimmunotherapy, (see, Potamianos et. al. Anticancer Res 20(2A):925-948 (2000)) for the detection of localized and metastatic cancers that exhibit expression of PSCA. Shedding or release of an extracellular domain of PSCA into the extracellular milieu, such as that seen for alkaline phosphodiesterase B10 (Meerson, N. Hepatology 27:563-568 (1998)), allows diagnostic detection of PSCA by anti-PSCA antibodies in serum andlor urine samples from suspect patients.
[0606] Anti-PSCA antibodies that specifically bind PSCA are used in therapeutic applications for the treatment of cancers that express PSCA. Anti-PSCA antibodies are used as an unconjugated modality and as conjugated form in which the antibodies are attached to one of various therapeutic or imaging modalities well known in the art, such as a prodrugs, enzymes or radioisotopes. In preclinical studies, unconjugated and conjugated anti-PSCA antibodies are tested for efficacy of tumor prevention and growth inhibition in the SCID mouse cancer xenograft models, kidney cancer models AGS-K3 and AGS-K6, (see, the Example entitled "PSCA Monoclonal Antibody-mediated Inhibition of Bladder and Lung Tumors In Vivo). Either conjugated and unconjugated anti-PSCA antibodies are used as a therapeutic modality in human clinical trials either alone or in combination with other treatments as described in following Examples.
Example 40: Human Clinical Trials for the Treatment and Diagnosis of Human Carcinomas through use of Human Anti-PSCA Antibodies In vivo [0607] Antibodies are used in accordance with the present invention which recognize an epitope on PSCA, and are used in the treatment of certain tumors such as those listed in Table I. Based upon a number of factors, including PSCA expression levels, tumors such as those listed in Table I are presently preferred indications. In connection with each of these indications, three clinical approaches are successfully pursued.
Adjunctive therapy: In adjunctive therapy, patients are treated with anti-PSCA antibodies in combination with a chemotherapeutic or antineoplastic agent and/or radiation therapy. Primary cancer targets, such as those listed in Table I, are treated under standard protocols by the addition anti-PSCA antibodies to standard first and second line therapy. Protocol designs address effectiveness as assessed by reduction in tumor mass as well as the ability to reduce usual doses of standard chemotherapy. These dosage reductions allow additional and/or prolonged therapy by reducing dose-related toxicity of the chemotherapeutic agent. Anti-PSCA antibodies are utilized in several adjunctive clinical trials in combination with the chemotherapeutic or antineoplastic agents adriamycin (advanced prostrate carcinoma), cisplatin (advanced head and neck and lung carcinomas), taxol (breast cancer), and doxorubicin (preclinical).
II.) Monotherapy: In connection with the use of the anti-PSCA antibodies in monotherapy of tumors, the antibodies are administered to patients without a chemotherapeutic or antineoplastic agent. In one embodiment, monotherapy is conducted clinically in end stage cancer patients with extensive metastatic disease. Patients show some disease stabilization. Trials demonstrate an effect in refractory patients with cancerous tumors.
IIl.) Imaging Agent: Through binding a radionuclide iodine or yttrium (1 131
Y
90 to anti-PSCA antibodies, the radiolabeled antibodies are utilized as a diagnostic and/or imaging agent. In such a role, the labeled antibodies localize to both solid tumors, as well as, metastatic lesions of cells expressing PSCA. In connection with the use of the anti-PSCA antibodies as imaging agents, the antibodies are used as an adjunct to surgical treatment of solid tumors, 119 WO 2005/014780 PCT/US2004/017231 as both a pre-surgical screen as well as a post-operative follow-up to determine what tumor remains andlor returns. In one embodiment, a (111 n)-PSCA antibody is used as an imaging agent in a Phase I human clinical trial in patients having a carcinoma that expresses PSCA (by analogy see, Divgi et al. J. Natl. Cancer Inst. 83:97-104 (1991)). Patients are followed with standard anterior and posterior gamma camera. The results indicate that primary lesions and metastatic lesions are identified.
Dose and Route of Administration [0608] As appreciated by those of ordinary skill in the art, dosing considerations can be determined through comparison with the analogous products that are in the clinic. Thus, anti-PSCA antibodies can be administered with doses in the range of 5 to 400 mg/m 2, with the lower doses used, in connection with safety studies. The affinity of anti- PSCA antibodies relative to the affinity of a known antibody for its target is one parameter used by those of skill in the art for determining analogous dose regimens. Further, anti-PSCA antibodies that are fully human antibodies, as compared to the chimeric antibody, have slower clearance; accordingly, dosing in patients with such fully human anti-PSCA antibodies can be lower, perhaps in the range of 50 to 300 mg/m 2 and still remain efficacious. Dosing in mg/m 2 as opposed to the conventional measurement of dose in mg/kg, is a measurement based on surface area and is a convenient dosing measurement that is designed to include patients of all sizes from infants to adults.
[0609] Three distinct delivery approaches are useful for delivery of anti-PSCA antibodies. Conventional intravenous delivery is one standard delivery technique for many tumors. However, in connection with tumors in the peritoneal cavity, such as tumors of the ovaries, biliary duct, other ducts, and the like, intraperitoneal administration may prove favorable for obtaining high dose of antibody at the tumor and to also minimize antibody clearance. In a similar manner, certain solid tumors possess vasculature that is appropriate for regional perfusion. Regional perfusion allows for a high dose of antibody at the site of a tumor and minimizes short term clearance of the antibody.
Clinical Development Plan (CDP) [0610] Overview: The CDP follows and develops treatments of anti-PSCA antibodies in connection with adjunctive therapy, monotherapy, and as an imaging agent, Trials initially demonstrate safety and thereafter confirm efficacy in repeat doses. Trails are open label comparing standard chemotherapy with standard therapy plus anti-PSCA antibodies. As will be appreciated, one criteria that can be utilized in connection with enrollment of patients is PSCA expression levels in their tumors as determined by biopsy.
[0611] As with any protein or antibody infusion-based therapeutic, safety concerns are related primarily to (i) cytokine release syndrome, hypotension, fever, shaking, chills; (ii) the development of an immunogenic response to the material development of human antibodies by the patient to the antibody therapeutic, or HAHA response); and, (iii) toxicity to normal cells that express PSCA. Standard tests and follov-up are utilized to monitor each of these safety concerns. Anti-PSCA antibodies are found to be safe upon human administration.
Example 41: Human Clinical Trial Adjunctive Therapy with Human Anti-PSCA Antibody and Chemotherapeutic Agent [0612] A phase I human clinical trial is initiated to assess the safety of six intravenous doses of a human anti-PSCA antibody in connection with the treatment of a solid tumor, a cancer of a tissue listed in Table I. In the study, the safety of single doses of anti-PSCA antibodies when utilized as an adjunctive therapy to an antineoplastic or chemotherapeutic 120 WO 2005/014780 PCT/US2004/017231 agent as defined herein, such as, without limitation: cisplatin, topotecan, doxorubicin, adriamycin, taxol, or the like, is assessed. The trial design includes delivery of six single doses of an anti-PSCA antibody with dosage of antibody escalating from approximately about 25 mg/m 2to about 275 mg/m 2 over the course of the treatment in accordance with the following schedule: DayO Day7 Day14 Day21 Day28 mAb Dose 25 75 125 175 225 275 mg/m2 mg/m2 mg/m2 mgfm 2 mg/m mg/m 2 Chemotherapy (standard dose) [0613] Patients are closely followed for one-week following each administration of antibody and chemotherapy. In particular, patients are assessed for the safety concerns mentioned above: cytokine release syndrome, hypotension, fever, shaking, chills; (ii) the development of an immunogenic response to the material development of human antibodies by the patient to the human antibody therapeutic, or HAHA response); and, (iii) toxicity to normal cells that express PSCA. Standard tests and follow-up are utilized to monitor each of these safety concerns. Patients are also assessed for clinical outcome, and particularly reduction in tumor mass as evidenced by MRI or other imaging.
[0614] The anti-PSCA antibodies are demonstrated to be safe and efficacious, Phase II trials confirm the efficacy and refine optimum dosing.
Example 42: Human Clinical Trial: Monotherapy with Human Anti-PSCA Antibody [0615] Anti-PSCA antibodies are safe in connection with the above-discussed adjunctive trial, a Phase II human clinical trial confirms the efficacy and optimum dosing for monotherapy. Such trial is accomplished, and entails the same safety and outcome analyses, to the above-described adjunctive trial with the exception being that patients do not receive chemotherapy concurrently with the receipt of doses of anti-PSCA antibodies.
Example 43: Human Clinical Trial: Diagnostic Imaqinq with Anti-PSCA Antibody [0616] Once again, as the adjunctive therapy discussed above is safe within the safety criteria discussed above, a human clinical trial is conducted concerning the use of anti-PSCA antibodies as a diagnostic imaging agent. The protocol is designed in a substantially similar manner to those described in the art, such as in Divgi et al. J. Natl. Cancer nst. 83:97- 104 (1991). The antibodies are found to be both safe and efficacious when used as a diagnostic modality.
Example 44 Homologv Comparison of PSCA v.4 to Known Sequences: [0617] The PSCA v.4 gene encodes a 189 aa protein. The human PSCA v.4 protein exhibit a high degree of homology to human prostate stem cell antigen (gi 27482160), exhibiting 98% identity to PSCA v.4 at the protein level (Figure The mouse homolog of PSCA v.4 has not been identified.
[0618] The PSCA v.4 protein has several variants (figure 11). These include 8 SNPs and a splice variant, referred to as PSCA v.3. The PSCA v.3 protein encompasses the C-termial portion of PSCA v.4, and corresponds to aa 94-189 of that variant. Bioinformatics analysis using topology prediction programs indicate that PSCA v.4 is a soluble protein with no transmembrane domains (Table WO 2005/014780 PCT/US2004/017231 [0619] Motif analysis revealed the presence of two protein functional motifs in the PSCA v.4 protein (Table L), namely a cadherin motif and a granulin domain have been identified. Cadherins belong to a family of calcium-dependent cell adhesion molecules. They are single transmembrane proteins containing immunoglobulin like domains, and are involved in cell adhesion and sorting (Shan et al, Biophys Chem 1999, 82:157). For examples, cadherins mediate tissuespecific cell adhesion of lymphocytes to the surface of epithelial cells. Cadherins have been shown to function in tissue morphogenesis, cell adhesion, cell differentiation, cell migration and tumour metastasis (Yap AS, Kovacs EM. J Biol Chem 2003, 160:11; Vestweber D. Curr Opin Cell Biol 2002,14:587; Bloom et al, Mol Biol Cell. 1999, 10:1521; Brodt P. Cancer Met Rev 1991, 10:23). Granulins or epithelins are growth factors originally purified from cell-conditioned media, shown to enhance cell proliferation (Xu, S. Q. et al, J. Biol. Chem. 1998, 273:20078). Granulins are expressed at elevated levels in several cancers, including gliomas and renal cancer (Liau L et al, Cancer Res. 60:1353, Donald, C. D et al, Anticancer Res. 21:3739).
[0620] The motifs found in PSCA v.4 indicate that PSCA v.4 can participate in tumor growth, and progression by regulating cell proliferation, cell adhesion, cell communication, invasion and metastasis.
[0621] Accordingly, when PSCA v.4 functions as a regulator of tumor establishment, tumor growth, tumor invasion, survival or cell signaling, PSCA v.4 is used for therapeutic, diagnostic, prognostic and/or preventative purposes. In addition, when a molecule, such as a splice variant or SNP of PSCA v.4 is expressed in cancerous tissues, such as those listed in Table I, they are used for therapeutic, diagnostic, prognostic and/or preventative purposes.
Example 45: Regulation of Transcription [0622] The mitochondrial localization of PSCA v.4 coupled to the presence of cadherin domains within its sequence indicates that PSCA v.4 modulates the transcriptional regulation of eukaryotic genes. Regulation of gene expression is confirmed, by studying gene expression in cells expressing or lacking PSCA v.4. For this purpose, two types of experiments are performed.
[0623] In the first set of experiments, RNA from parental and PSCA v.4-expressing cells are extracted and hybridized to commercially available gene arrays (Clontech) (Smid-Koopman E et al. Br J Cancer. 2000. 83:246). Resting cells as well as cells treated with FBS, androgen or growth factors are compared. Differentially expressed genes are identified in accordance with procedures known in the art. The differentially expressed genes are then mapped to biological pathways (Chen Ket al. Thyroid. 2001. 11:41.).
[0624] In the second set of experiments, specific transcriptional pathway activation is evaluated using commercially available (Stratagene) luciferase reporter constructs including: NFkB-luc, SRE-luc, ELK1-luc, ARE-luc, p53-luc, and CREluc. These transcriptional reporters contain consensus binding sites for known transcription factors that lie downstream of well-characterized signal transduction pathways, and represent a good tool to ascertain pathway activation and screen for positive and negative modulators of pathway activation.
[0625] Thus, PSCA v.4 plays a role in gene regulation, and it is used as a target for diagnostic, prognostic, preventative and/or therapeutic purposes.
Example 46: Identification and Confirmation of Potential Signal Transduction Pathways [0626] Many mammalian proteins have been reported to interact with signaling molecules and to participate in regulating signaling pathways. (J Neurochem. 2001; 76:217-223). Cadherin molecules have been associated with Cdc42 122 WO 2005/014780 PCT/US2004/017231 and Rho signaling (Kouklis J Biol Chem. 2003, 278: 16230). Using immunoprecipitation and Western blotting techniques, proteins are identified that associate with PSCA v.4 and mediate signaling events. Several pathways known to play a role in cancer biology can be regulated by PSCA v.4, including phospholipid pathways such as P13K, AKT, etc, adhesion and migration pathways, including FAK, Rho, Rac-1, catenin, etc, as well as mitogenic/survival cascades such as ERK, p38, etc (Cell Growth Differ. 2000,11:279; J Biol Chem. 1999, 274:801; Oncogene. 2000,19:3003, J. Cell Biol. 1997,138:913.).
In order to determine whether expression of PSCA v.4 is sufficient to regulate specific signaling pathways not otherwise active in resting cancer cells, the effect of PSCA v.4 on the activation of the signaling cascade is investigated in the cancer cell lines PA-1, Panel and Daudi. Cancer cells stably expressing PSCA v.4 or neo are stimulated with growth factor, FBS or other activating molecules. Whole cell lysates are analyzed by western blotting.
[0627] To confirm that PSCA v.4 directly or indirectly activates known signal transduction pathways in cells, luciferase (luc) based transcriptional reporter assays are carried out in cells expressing individual genes. These transcriptional reporters contain consensus-binding sites for known transcription factors that lie downstream of vellcharacterized signal transduction pathways. The reporters and examples of these associated transcription factors, signal transduction pathways, and activation stimuli are listed below.
1. NFkB-luc, NFkB/Rel; Ik-kinase/SAPK; growthlapoptosis/stress 2. SRE-luc, SRF/TCF/ELK1; MAPK/SAPK; growth/differentiation 3. AP-1-luc, FOS/JUN; MAPK/SAPK/PKC; growth/apoptosis/stress 4. ARE-luc, androgen receptor; steroids/MAPK; growth/differentiation/apoptosis p53-luc, p53; SAPK; growth/differentiation/apoptosis 6. CRE-luc, CREB/ATF2; PKA/p38; grovth/apoptosis/stress 7. TCF-luc, TCF/Lef; 0-catenin, Adhesion/invasion [0628] Gene-mediated effects can be assayed in cells showing mRNA expression. Luciferase reporter plasmids can be introduced by lipid-mediated transfection (TFX-50, Promega). Luciferase activity, an indicator of relative transcriptional activity, is measured by incubation of cell extracts with luciferin substrate and luminescence of the reaction is monitored in a luminometer.
[0629] Signaling pathways activated by PSCA v.4 are mapped and used for the identification and validation of therapeutic targets. When PSCA v.4 is involved in cell signaling, it is used as target for diagnostic, prognostic, preventative and/or therapeutic purposes.
Example 47: Involvement in Tumor Progression [0630] Based on the role of granulin and cadherin motifs in cell growth, adhesion and protein interactions, the PSCA v.4 gene can contribute to the growth, adhesion, invasion and transformation of cancer cells. The role of PSCA v.4 in tumor growth is confirmed in a variety of primary and transfected cell lines including prostate cell lines, as well as NIH 3T3 cells engineered to stably express PSCA v.4. Parental cells lacking PSCA v.4 and cells expressing PSCA v.4 are evaluated for cell growth using a well-documented proliferation assay (Fraser SP, Grimes JA, Djamgoz MB. Prostate.
2000;44:61, Johnson DE, Ochieng J, Evans SL. Anticancer Drugs. 1996, 7:288).
[0631] To confirm the role of PSCA v.4 in the transformation process, its effect in colony forming assays is investigated. Parental NIH-3T3 cells lacking PSCA v.4 are compared to NIH-3T3 cells expressing PSCA v.4, using a soft agar assay under stringent and more permissive conditions (Song Z. et al. Cancer Res. 2000;60:6730).
WO 2005/014780 PCT/US2004/017231 [0632] To confirm the role of PSCA v.4 in invasion and metastasis of cancer cells, a well-established assay is used, a Transwell Insert System assay (Becton Dickinson) (Cancer Res. 1999; 59:6010). Control cells, including prostate, pancreas and kidney cell lines lacking PSCA v.4 are compared to cells expressing PSCA v.4. Cells are loaded with the fluorescent dye, calcein, and plated in the top well of the Transwell insert coated with a basement membrane analog.
Invasion is determined by fluorescence of cells in the lower chamber relative to the fluorescence of the entire cell population.
[0633] PSCA v.4 can also play a role in cell cycle and apoptosis. Parental cells and cells expressing PSCA v.4 are compared for differences in cell cycle regulation using a well-established BrdU assay (Abdel-Malek ZA. J Cell Physiol.
1988, 136:247). In short, cells are grown under both optimal (full serum) and limiting (low serum) conditions are labeled with BrdU and stained with anti-BrdU Ab and propidium iodide. Cells are analyzed for entry into the G1, S, and G2M phases of the cell cycle. Alternatively, the effect of stress on apoptosis is evaluated in control parental cells and cells expressing PSCA v.4, including normal and tumor prostate cells. Engineered and parental cells are treated with various chemotherapeutic agents, such as etoposide, taxol, etc, and protein synthesis inhibitors, such as cycloheximide. Cells are stained with annexin V-FITC and cell death is measured by FACS analysis. The modulation of cell death by PSCA v.4 can play a critical role in regulating tumor progression and tumor load.
[0634] When PSCA v.4 plays a role in cell growth, transformation, invasion or apoptosis, it is used as a target for diagnostic, prognostic, preventative and/or therapeutic purposes.
Example 48: Involvement in Angiogenesis [0635] Angiogenesis or new capillary blood vessel formation is necessary for tumor growth (Hanahan D, Folkman J.
Cell. 1996, 86:353; Folkman J. Endocrinology. 1998 139:441). Several assays have been developed to measure angiogenesis in vitro and in vivo, such as the tissue culture assays endothelial cell tube formation and endothelial cell proliferation. Using these assays as well as in vitro neo-vascularization, the role of PSCA v.4 in angiogenesis, enhancement or inhibition, is confirmed.
[0636] For example, endothelial cells engineered to express PSCA v.4 are evaluated using tube formation and proliferation assays. The effect of PSCA v.4 is also confirmed in animal models in vivo. For example, cells either expressing or lacking PSCA v.4 are implanted subcutaneously in immunocompromised mice. Endothelial cell migration and angiogenesis are evaluated 5-15 days later using immunohistochemistry techniques. PSCA v.4 affects angiogenesis, and it is used as a target for diagnostic, prognostic, preventative andfor therapeutic purposes.
Example 49: Involvement in Protein-Protein Interactions [0637] Cadhesrin motifs have been shown to mediate interaction with other proteins. Using immunoprecipitation techniques as well as two yeast hybrid systems, proteins are identified that associate with PSCA v.4. Immunoprecipitates from cells expressing PSCA v.4 and cells lacking PSCA v.4 are compared for specific protein-protein associations.
[0638] Studies are performed to confirm the extent of association of PSCA v.4 with effector molecules, such as nuclear proteins, transcription factors, kinases, phosphates etc. Studies comparing PSCA v.4 positive and PSCA v.4 negative cells as well as studies comparing unstimulated/resting cells and cells treated with epithelial cell activators, such as cytokines, growth factors, androgen and anti-integrin Ab reveal unique interactions.
[0639] In addition, protein-protein interactions are confirmed using two yeast hybrid methodology (Curr Opin Chem Biol. 1999, 3:64). A vector carrying a library of proteins fused to the activation domain of a transcription factor is introduced 124 WO 2005/014780 PCT/LS2004/017231 into yeast expressing a PSCA v.4-DNA-binding domain fusion protein and a reporter construct. Protein-protein interaction is detected by colorimetric reporter activity. Specific association with effector molecules and transcription factors directs one of skill to the mode of action of PSCA v.4, and thus identifies therapeutic, prognostic, preventative and/or diagnostic targets for cancer. This and similar assays are also used to identify and screen for small molecules that interact with PSCA v.4.
[0640] Thus it is found that PSCA v.4 associates with proteins and small molecules. Accordingly, PSCA v.4 and these proteins and small molecules are used for diagnostic, prognostic, preventative and/or therapeutic purposes.
Example 50: Involvement of PSCA v.4 in cell-cell communication [0641] Cell-cell communication is essential in maintaining organ integrity and homeostasis, both of which become deregulated during tumor formation and progression. Based on the presence of a cadhesrin motif in PSCA v.4, a motif known to be involved in cell interaction and cell-cell adhesion, PSCA v.4 can regulate cell communication. Intercellular communications can be measured using two types of assays Biol. Chem. 2000, 275:25207). In the first assay, cells loaded with a fluorescent dye are incubated in the presence of unlabeled recipient cells and the cell populations are examined under fluorescent microscopy. This qualitative assay measures the exchange of dye between adjacent cells. In the second assay system, donor and recipient cell populations are treated as above and quantitative measurements of the recipient cell population are performed by FACS analysis. Using these two assay systems, cells expressing PSCA v.4 are compared to controls that do not express PSCA v.4, and it is found that PSCA v.4 enhances cell communications. Small molecules and/or antibodies that modulate cell-cell communication mediated by PSCA v.4 are used as therapeutics for cancers that express PSCA v.4. When PSCA v.4 functions in cell-cell communication and small molecule transport, it is used as a target or marker for diagnostic, prognostic, preventative and/or therapeutic purposes.
[0642] Throughout this application, various website data content, publications, patent applications, and patents are referenced. (Websites are referenced by their Uniform Resource Locator, or URL, addresses on the World Wide Web.) The disclosures of each of these references are hereby incorporated by reference herein in their entireties. In addition, this application relates to U.S. Serial No. 091359,326, filed July 20, 1999; U.S. Serial No. 09/308,503, filed May 25, 1999; U.S.
Serial No. 091251,835, filed February 17, 1999; U.S. Serial No. 09/203,939, filed December 2, 1998; U.S. Serial No.
091038,261, filed March 10, 1998; U.S. Serial No. 081814,279, filed March 10, 1997; U.S. Serial No. 60/071,141 filed January 12, 1998; U.S. Serial No. 60! 074,675, filed February 13, 1998; U.S. Serial Nos. 601124,658, filed March 16,1999; U.S. serial No. 601120,536 filed February 17, 1999; and 60/113,230 filed December 21, 1998. The contents of all of the foregoing applications are fully incorporated by reference into the present application.
[0643] The present invention is not to be limited in scope by the embodiments disclosed herein, which are intended as single illustrations of individual aspects of the invention, and any that are functionally equivalent are within the scope of the invention. Various modifications to the models and methods of the invention, in addition to those described herein, will become apparent to those skilled in the art from the foregoing description and teachings, and are similarly intended to fall within the scope of the invention. Such modifications or other embodiments can be practiced without departing from the true scope and spirit of the invention.
WO 2005/014780 WO 205/04780PCT/1§S2004/017231
TABLES:
TABLE 1: Tissues that Express PSCA: a. Malicinant Tissues Prostate Pancreas Bladder Kidney Colon Lung Ovary Breast b. Normal Tissues TABLE 11: Amino Acid Abbreviations SINGLE LETTER THREE LETTER FULL NAME F Phe phenylalanine L Leu leucine S Ser serine Y Tyr tyrosine C Cys cysteine W Trp tryptophan P Pro proline H His histidine Q Gin glutamine R Arg arginine Ilie isoleucine M Met methionine T Thr threonine N Asn asparagine K Lys lysine V Val valine A Ala alanine D Asp aspartic acid E Glu glutamnic acid G Gly glycine WO 2005/014780 PCT/US2004/017231 TABLE III: Amino Acid Substitution Matrix Adapted from the GCG Software 9.0 BLOSUM62 amino acid substitution matrix (block substitution matrix). The higher the value, the more likely a substitution is found in related, natural proteins. (See world wide web URL ikp.unibe.ch/manual/blosum62.html A C D E F G H I K L M N P Q R S T V W Y.
4 0 -2 -1 -2 0 -2 -1 -1 -1 -1 -2 -1 -1 -1 1 0 0 -3 -2 A 9 -3 -4 -2 -3 -3 -1 -3 -1 -1 -3 -3 -3 -3 -1 -1 -1 -2 -2 C 6 2 -3 -1 -1 -3 -1 -4 -3 1 -1 0 -2 0 -1 -3 -4 -3 D -3 -2 0 -3 1 -3 -2 0 -1 2 0 0 -1 -2 -3 -2 E 6 -3 -1 0 -3 0 0 -3 -4 -3 -3 -2 -2 -1 1 3 F 6 -2 -4 -2 -4 -3 0 -2 -2 -2 0 -2 -3 -2 -3 G 8 -3 -1 -3 -2 1 -2 0 0 -1 -2 -3 -2 2 H 4 -3 2 1 -3 -3 -3 -3 -2 -1 3 -3 -1 I -2 -1 0 -1 1 2 0 -1 -2 -3 -2 K 4 2 -3 -3 -2 -2 -2 -1 1 -2 -1 L -2 -2 0 -1 -1 -1 1 -1 -1 M 6 -2 0 0 1 0 -3 -4 -2 N 7 -1 -2 -1 -1 -2 -4 -3 P 1 0 -1 -2 -2 -1 Q -1 -1 -3 -3 -2 R 4 1 -2 -3 -2 S 0 -2 -2 T 4 -3 -1 V 11 2 W 7 Y WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLE IV: HLA Class IM1 MotifslSupermotifs TABLE IV HLA Class I SupermotifslMotifs SUPERMOTIF POSITION POSITION POSITION 2 (Primary Anchor) 3 (Primary Anchor) C Terminus (Primary Anchor) Al TILVMS FWY A2 LIVMATQ IVMATL A3 VSMATLI RK A24 YFWIVLMT FIYWLM 137 P VILFMWYA B27 RHK FYLWMIVA B44 ED FWYLIMVA B58 ATS FWYLIVIAA B62 QLIVMP FWYMIVLA
MOTIFS
Al TSM Y Al IDEAS Y A2.1 LMVQIAT VLIMAT A3 LMVISATFCGD KYRHFA All VTMLISAGNCDF KRYH A24 YFWM FLIW A*31 01 MVTALIS RK A*3301 MVALFIST RK A*6801 AVTMSLI RK B*0702 P LMFW'(AIV B3*3501 P LMFWVYiVA B51 P LIVFWYAM B"5301 P IMFWYALV 93*5401 P ATIVLMFWY Bolded residues are preferred, italicized residues are less preferred: A peptide is considered motif-bearing if it has primary anchors at each primary anchor position for a motif or supermotif as specified in the above table.
TABLE IV HLA Class 11 Supermotif 1 6 9 W, FY,V,.I, L A, V,,LP, C,S, T A, V,1, L, C,S, T,M, Y WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLE IV HLA Class 11 Motifs MOTIFS 10* anchor I 2 3 4 5 1l'anchor 6 7 8 9 DR4 preferred FMYLiVW M T I VSTCPALJM MH NAH deleterious W R WIDE DRI preferred MFLIVWY PAMQ VMATSPLIC M AVM deleterious GCOH ID CWID GDE D DR7 preferred MFLIVWY M W A IVMSACTPL M IV deleterious C G GRID N G DR3 MOTIFS V anchorl1 2 3 1' anchor4 5 10' anchor 6 Motif a preferred LIVMFY D Motif b preferred LIVMFAY DNQEST KRH DR Supermotif MFLIVWY VMSTACPLI Italicized residues indicate less preferred or "tolerated' residues TABLE IV HIA Class I Supermotis POSITION: 1 2 3 4 5 6 7 8 C-terminus
SUPER-
MOTIFS
Al 1* Anchor 10 Anchor TIL MS
FWY
A2 10 Anchor 10* Anchor LIVMATQ
LIVMAT
A3 Preferred 10 Anchor YFW YFW YFW P 10 Anchor VSMATLI
RK
deleterious DE DE P A24 10 Anchor 10 Anchor YFW! VLMT lY L 137 Preferred FWY (515), 10Anchor FWY FWY I 0 Anchor LIVM P (415) VILFMWYA deleterious DE DE G ON DE (415) QN(315) B27 10 Anchor 1l 0 Anchor RI-K
FYLWMIVA
B44 V 0 Anchor 1V Anchor ED
FWYLIMVA
B58 1' Anchor 1 0 Anchor ATS
FWYLIVMA
B62 10' Anchor 10* Anchor QLIVMP FlWYMIVLA Italicized residues indicate less preferred or "tolerated" residues WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEF IV HLA Class I Motifs POSITION I 3 4 5 6 7 8 9 0terminus or C-terminus Al preferred GFYW 1 0 Anchor DEA YEW P DEQN YEW I 0 Anchor 9-mer STM
Y
deleterious DE RHKLIVMP A G A Al preferred GRHK ASTCUIVM loAnchor GSTC ASTO LIVM DE 1 0 Anchor 9-mer DEAS
Y
deleterious A RHKDEPYFW DE PQN RHK PG GP Al preferred YEWV I 0 Anchor DEAQN A YFWVQN PASTC GDE P 1 *Anchor STM
Y
mer deleterious GP RHKGLIVM DE RHK QNA RHKYFW RHK A Al preferred YEWV STCLIVM 1 0 Anchor A YEWV PG G YEW I 0 Anchor DEAS
Y
mer deleterious RI-K RHKDEPYFW P G PRHK QN A2.1 preferred YEW I 'Anchor YEW STC YEW A P l 0 Anchor 9-mer LMIVQAT
VLIMAT
deleterious DEP DERKH RKH DERKH POSITION:l1 2 3 4 5 6 7 8 9 0- Terminus A2.1 preferred AYFW 1 *Anchor LVIM G G FYWL I 0 Anchor LMIVQAT vim VLIAT mer deleterious DEP DE RKHA P RKH DERKHRI H A3 preferred RHK I 0 Anchor YEWV PRHKYF A YFW P 1 0 Anchor LMVISATECGD W
KYRHFA
deleterious DEP DE All preferred A 1 'Anchor YEWV YEW A YEW YEW P 1 0 Ancher VTLMISAGN CD
KRYH
F
deleterious DEP A G A24 preferred YFWRI-K l*Anchor STC YEW YEW l 0 Anchor 9-mer YFWA4
FLIW
deleterious DEG DE G QNP DERHKG AQN A24 Preferred I 0 Anchor P YEWP P 1 0 Anchor YFWM EL1W mer Deleterious GDE QN RHK DE A QN DEA RHK l'Anchor YEW P YEW YEW AP l 0 Anchor MVTALIS
RK
Deleterious DEP DE ADE DE DE DE A33C1 Preferred I 0 Anchor YEW AYEW I *Anchor MVALFIST
RK
Deleterious GP DE A6801 Preferred YFWSTC l'Anchor YEWLIV YEW P I *Anchor AVTMSL/ M RK deleterious GP DEG RHIK A B0702Preferred RHKFWY I 0 Anchor RHK RHK RHK RHK PA I -Anchor P
LMEWYAI
V
WO 2005/014780 WO 205/04780PCT/1§S2004/017231 POSITION I 3 4 5 6 7 8 9 C..
terminus Al preferred GEYW I Anchor IDEA YFW 9-mer STM deleterious DE RHKLIVMP A Al preferred GRHK ASTCLIVM l 0 Anchor GST( 9-mer DEAS deleterious A RHKIJEPYFW DE deleterious DEQNP DEP DE 83501 Preferred FWYLIVM I Anchor EWY
P
deleterious AGP B51 Preferred LIVMFWY 1 'Anchor FYVY STC
P
deleterious AGPDER
HKSTC
B5301 preferred LIVMFWY 1 0 Anchor EWY STO
P
or C-terminus P IDEQN YEW I *Anchor
Y
G A ASTC LIVM DE 1 0 Anchor
Y
PQN RHK PG GP DE GDE QN DE EWY l 0 Anohor LMF WY/V
A
G G FWVY G FWY 1 'Anchor LIVE WYA
M
DE G DEQIN GDE FWVY LIVMFWYF\NY l 0 'Anchor
IMEWYAL
V
G RHKQN DE LIVM ALIVM FWYA l 0 Anchor P ATIVLA4F
WY
RHKDE DE QNDGE DE deleterious AGPQN B5401 preferred EVFY 1 Anchor
P
deleterious GPQNDE
EWYLIVM
GDESTC
WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLE IV Summary of HILA-supertypes Overall phenotypic frequencies of H LA-supertypes in different ethnic populations ____Specificity _____Phenotypic frequeny,,,,,,,, Supertype Position 2 IC-Terminuc Caucasiar. N.A. Black Japanese Chinese Hispnic eag B7 ILMVFWY43.2 55.1 57.1 43.0 49.3 49.5 A3 AILMVST RK _37.5 42.1 45.8 52.7 43.1 4.2 A2 AILMVT 6ILMVT 45.8 39.0 42.4 45.9 43.0 42.2 A24 YF (WIVLMT)IFI (YWLM) 23,9 38.9 58.6 40.1 38.3 40.0 B44 Ej(D) IFWYLIMVA43,0 21.2 42.9 39.1 39.0 37.0 1 TI (LVMS) FWY 47.1 16.1 21.8 14.7 26.3 25.2 B27 RHK FYL (WMI) 28.4 26.1 13.3 13.9- 35. 3 23.4 B62 QL (IVMP) FNY (MIV) 12.6 4.8 36.5 25.4 11.1 18.1 8B58 ATS FVY (UI) ;10.0 251 16 90 .9 03 TABLE IV ,i 0 *d n covra~ e fforde.d hy different HLA-sunertvo~e combinations HLA-supertypes Phenotypic frequency Caucasian INA Blacks Japanese Ciee Hispanic Avera e,, 83.0 86.1 87.5 88.4 86.3 86.2 A2, A3 and B7 99.5 98.1 100.0 99.5 9.4 99.3 A2, A3, B7, A24, 99.9 99.6 100.0 99.8 99.9 99.8 B44 and Al ~A2, A3, B7, A24, 1B44, Al, B27, B62, and B 58 tiotifs indicate the residues defining supertype specificites. The motifs incorporate residues determined on the basis or )ubl!Ehed data to be recognized by multiple alleles within the supertype. Residues within brackets are additional residues also predicted to be tolerated by multiple alleles within the supertype.
Table V: Frequently Occurring Motifs Name avrg. Description Potential Function Nucleic acid-binding protein functions as transcription factor, nuclear location zf-C2H2 34% Zinc finger, C2H-2 type probable Cytoobrome b(N- membrane bound oxidase, generate c tochrome-bN 68% term inal)/b6/petB superoxide domains are one hundred amino acids long and include a conserved Ig -19% Immunoglobulin domain intradomain disulfide bond.
tandem repeats of about 40 residues, each containing a Tmp-Asp motif.
Function in signal transduction and 18% WD domain, G-beta repeat protein interaction may function in targeting signaling PDZ 23% PDZ domain molecules to sub-membranous sites LRR 28% Leucine Rich Repeat short sequence motifs involved in protein-protein interactions WO 2005/014780 PCT/US2004/017231 conserved catalytic core common to both serine/threonine and tyrosine protein kinases containing an ATP Pkinase 23% Protein kinase domain binding site and a catalytic site pleckstrin homology involved in intracellular signaling or as constituents PH 16% PH domain of the cytoskeleton 30-40 amino-acid long found in the extracellular domain of membrane- EGF 34% EGF-like domain bound proteins or in secreted proteins Reverse transcriptase (RNA-dependent DNA Rvt polymerase) Cytoplasmic protein, associates integral Ank 25% Ank repeat membrane proteins to the cytoskeleton NADH- membrane associated. Involved in Ubiquinone/plastoquinone proton translocation across the Oxidored_ql 32% (complex various chains membrane calcium-binding domain, consists of a12 residue loop flanked on both sides by a Efhand 24% EF hand 12 residue alpha-helical domain Retroviral aspartyl Aspartyl or acid proteases, centered on Rvp 79% protease a catalytic aspartyl residue extracellular structural proteins involved in formation of connective tissue. The Collagen triple helix repeat sequence consists of the G-X-Y and the Collagen 42% (20 copies) polypeptide chains forms a triple helix.
Located in the extracellular ligandbinding region of receptors and is about 200 amino acid residues long with two pairs of cysteines involved in disulfide Fn3 20% Fibronectin type Ill domain bonds seven hydrophobic transmembrane regions, with the N-terminus located 7 transmembrane receptor extracellularly while the C-terminus is 7tm 1 19% (rhodopsin family) cytoplasmic. Signal through G proteins Table VI: Post-translational modifications of PSCA v.4 N-glycosylation site 91 94 NASL (SEQ ID NO:147) cAMP- and cGMP-dependent protein kinase phosphorylation 13 RRTS (SEQ ID NO:148) Protein kinase C phosphorylation site 2-4 THR 12-14 TSR 37 SLR 51 53 SYR N-myristoylation site 120 125 GSIDTD (SEQ ID NO:149) Proline-rich region 18-134 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Table VII: Search Peptidles Variant 1 aa 1-123: 9-mers, 1 0-mers, 15-mers (SEQ ID NO: 150) MKA'VLLALLM AGLALQPGTA LLCYSCKAQV SNEDCLQVEN CTQLG3EQCWT ARIRAVGLLT VISKGCSLNC VDDSQDYYVG KKNITCCDTD LCNASGAHAL QPAAAILALL 2ALGLLLWGP 120 GQL 123 v.4 aa 1-189: 9-mers, 1lO-mers, 15-mers (SEQ ID NO: 151) MTHRTPTTqAR RTSRAV'TTC ATPAGPMPCS RLPSLRCSL HSACCSGDPA SYRLWGAPLQ PTLGVVPQAS VPLLTHPAQW EPVLVPEAHP NASLTMYVCA PVPHPDPP4A LSRTPTRQIG 120 SIDTDPPADG PSNPLCCCFHI GPAFSTLNPV LRHLFQEAF PAHPIYDLSQ VWSVVSPAPS 180 RGQALRAR 189 PSCAv.19 9-mers aa 25-41 GPMPCSRLLPSLRCSLH (SEQ ID NO:152) IG-mers aa24-42 AGPL4PCSRLLPSLRCSLHS (SEQ ID NO:153) aa 19-47 TCATPAGPMPCSPRLLPSLRCSLHiSACCSG (SEQ ID NO:154) PSCA 9-mers aa 44-60 CCSGDPASSRLWGAPLQ (SEQ ID NO:155) I 0-mers aa 43-6 1 ACCSGDPASSRLWGALDLQP (SEQ ID N0156) 1 5-mers aa 38-66 CSLHSACCSGDEASSRLWGAPLQPTLGVV (SEQ ID NO:157) PSCA v.21 9-mers aa 68-84 OASVPLLTDPAQWEPVL (SEQ ID NO:158) aa 67-85 FQASVPLLTDPAQWEPVLV (SEQ ID NO:159) aa 62-90 TIAGVVPQASVPLLTDPA7QWEPVLVL:EAHP (SEQ ID NO:160) PSCA v.21122 9-mers aa 69-84 ASVPLLTDLAQWEPVL (SEQ ID NO:161) I 0-mers aa QASVPLLTDLAQWEPVLV (SEQ ID NO:162) aa 63-90 LGVVPQASVPLLTDLAQWFPVLVPEAHP (SEQ ID NO:163) PSCA v.22 9-mers aa 69-85 ASVPLLTHLAQWZPVLV (SEQ ID NO:1 64) I 0-mers aa 68-86 QASVPLLTHLAQWEPVLVpI (SEQ ID NO: 165) 1 5-mors aa 63-91 LGVVPQASVPLLTHLAQWEPVLVFEAHFN (SEQ ID NO:166) PSCA v.24 9-mers aa 92-1 08 ASLTMYVCTPVPHPDP (SEQ ID NO:167) WO 2005/014780 WO 205/04780PCT/1§S2004/017231 1lO-mers aa 91-109 NASLTMYVCTVPHBDPPM (SEQ ID NO:1 68) aa 96-114 PEAHPNASLT4YVCTPVPH-PDPPMALSRT (SEQ ID NO:169) PSCA 9-mers aa 112-128 SRTPTRQISSIDTDPPA (SEQ ID NO:170) 1lO-mers aa 111-129 LSRTPTRQISSIDTDPPAD (SEQ ID NO:171) aa 106-1 34 DPPMALiSRTPTRQISSIDTDPIPADGP:)SNP (SEQ ID NO:172) PSCAv.25126 9-mers aa 27-128 TPTRQISSSDTDPPA (SEQ ID NO:173) 1 0-mers aa 26-1 29 RTPTRQISSSDTDPPAD (SEQ ID NO:174) 1 5-mors aa 108-1 34 Pb4ALSRTPTRQISSSDTDPPADGPSNP (SEQ ID NO:176) PSCA v.26 9-mers aa 114-130 TPTRQIGSSDTDPPADG (SEQ ID NO:176) aa 113-131 RTPTRQIGSSDTDPPADGP (SEQ ID NO:177) I 5-mers aa 108-136 PMALSRTPTRQIGSSDTDPPADGPSNPLC (SEQ ID NO:178) PSCA v.27 9-mers aa 181 -189 RGQALRRAQ (SEQ IDNIO:179) IlO-mers aa 180-1 89 SRGQALRRAQ (SEQ ID NO:180) aa 175-189 VSPAPSRGQALRRAQ (SEQ ID NO:1 81) WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Tables ViII TABLEVIII-VI-HLA-AI -9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9, amino acids, and the end position for each peptide is the start ____position pl us ei'ght.
1F707 [VDSQDYY 1[25=.000 IL4 199~ i.50 [J1 ERi QGTALLCY [_0..625 11 DSQDYYVGK R] 0.600 CDDSQY RE0.500 86' CC...N Rk.
56 J[ -VGLLTVISK ]1,0.250 IF 14 7iF ALQPGTALL RE0.-2-00 I 19- T AL L CY SC-K F0.200 F I A L LP AL G LL T1q.-9-9 FVSNEDCLQV 10,075 1 TpTLCN ',14o 17 LALLPALGIL.' 0.050 2 fKVLLALLM R[9 q AVLLA ILLMA. JRE -05 I 14 I AILALLPA -oo F717 Iv- VDDYrE Too IF -VLLALLMAG 5 .050 -7 FALLMAGLAL 0.5 LLPALGLL 0.05 18 ITCCDTDLC 0.02 if 411CqiLGQC 0.25 I 59 i TV[SKGCS F0.025 22 J LYSCp-K P I161ILALLPALG .02 .8 .LMAGLALQ 0.020 LTALVIII-V-HLA-A-9MERS- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
ISTRTSUBSEQUENCE1 SCORE [7 =E LLALLMAGL ]10.02 j8,F HALQPAAAI ]F 0.020 I105 4AILALLPAL Ipoo 96 E GAHALQPAA]RE oK K3 NASGAHALQ. F5oo [114F-- 4GLLGPQ .020 I 66 CSNCVDDS -r0.015 114QPAAAILAL T -0.013 libq P PLGLLWV .1 ~Ts 41 FLLLWGPG16EF6 j13 E LALQPGTAL F0.010, 27' [K EDCFi-0010 F.20_ [F ALLCKA ,0.010 4 C EcTO 0.010 1 QLGEQCWTA 0,010 F ~4~vNEDC FQ05010 I TE.9LQVNCTF 01 E77[AV LWV 0.010 59 4NCTQLGEQCF0.010 GTA_ .1 F83F N1TCCDTDLI '0.010 76 LLMAGLA 0 F72 LLCYSCKAQ 0.010 60 4 TVISKGCSL 0.010 67 [SLNCVD 1-1 FAG~Q~I 0,01 57~ F97 GLTVSK 001 162 i I S t.LN 5E0Yoa TABLEVIII14-ILA-All-9MERS-
PSCA
Each peptide is a portion of SEQ1 ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
iF7E7 LQPAAAILA 0.007 7 QCWARI FRIRAVGLLT .i 0.005 103 FAAAILALLP Loco 24 j YSCKAQVSN Fo.0003 F 1 NC-TQ LEqk 0003-L 113 'FLLLWGPG 00 1 Fo_ 1SAHLPA10 3 9 I LMAGLALQP Yp.O 7 '92 ONASGAHAL 0P9 53' 1IRAVG LLTV 0.0 I L TIRGL 0002 102_ PAAAILALL 1I0,002j 1 .5 F G6SLNCVDD_1 0.002 42 T71QLGEQW 0. 6 67 2 QF9 NDCL Ko6.6092 1-3,6 KLQVENCTQK 10. 002,, I11,2 ALGLLLWVGP 0 .0077 'F45 FGEQCWTARI 176 oi [I77 DDSQDYYVG 1 001 ,IY -F C-9QPmAF Iooo js VISKGCSLNI .0 48 CWTARIRAV 0.001q 11 KKNITCCDT 0.aoi WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEVIII-V1-HLA-A1 -9MERS-
PSCA
Each peptide is a portion of SEQI ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position position plus eight.___ FSTARaT SUBSEqUENCE; SCORE: 89 TDLCNASGA .0 TABLEVUII-V4-H LA-Al -9MERS-
___PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position! *plus eight, START, SBSEQUENCE SCOE: [i 15Ci EFPAHPIY 1000 46 SGD)PASYRL 1 2.500 104 IHPDPPMALS .12.500 123 IDD P 1.250 CCiISGDPASYifl000 14 WI FSTLNPVLR ~[0.300 8 VPEAHPNAS [0,225, 100 MALSRTPTR if0,200 136.i CCCFHGPAF 0.200 [7371, SVVSPAPSR f0.200 11 PSNPL CCCF 0.15 8j9 f, HPNASLTMY F-9.11 D113[ IRTPTRQIGS 0.125 14 [STLNPVLRH 0.125 F8 j VLVPAHPN f0.100 1 7 RLPPSLRCS .1 100 8r [AHIPNASLT 0.100 1467 ILNPVLRHL 0.100 1 21 SIDIOPPAD Fb.T0I 7F 9: qkWEPVLVPE i0.090 f185 IYDLSQVWS 0.05 L YCAPV 005 TALEVI II-V4-H LA-Al -9MERS-
PSCA
Each peptidle is a portion of SEQ IDi NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start posto plus eight. S ARlI JSUBSEQUENCE SCORE! ,F 1277 PAGPSNPI L 0.050 J102 j[ -VPHPDPPMA i 1 0.060 55 FjWGAPLQPTL
F*-
'7 L QASVPLLTH IF o.:O 5 7 92] ASILTMY A [0.030 1J68 LS§QVWSVVS 0.030 11j17271~ WSVSAS[ 1O.o3o 0 jlI SILPPSLRC Q.925 47i LNPVLRHLF_'7 0 6-L 1=176 SPAPSRGQA I 0.0o25 16 Y PTATPA FO [0025 '74 ~lLTHPAQWEP i[0.025: F4 RTTTWARRT O0025 5F-- TTTWARRTS 0F.0-25 .f I NASLTMYV 020: F1~ 43 C sG 0.020 3RSLHSACC 0O.020 1 AVTPTCATP 000 jCATPAGPMP 10.020 LV7~PEAHPNA .2 [65 PASLf002 I 3 CCFHGPAFS 7.2 'I13IHLFPQEAFP 00 2:97 I qSRLPPSLR 0O.015 6 9- A'-s v PLLTH P F6.01i5 [175 i VSPAlPSRGQ 0.015 38 CSLHSACCS OQi _Qi GPSNPLCC 0.0137.
]IDPASYRLWG 1 0.013' T57 I1 APLQPTLGV 10.013 TBLEVIII-V4-HLA-A1-9MERS-
PSCA
Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
STARTI SUBSEQUENCEJ
SCORE
I 1817. PAHPIYDLSi01 L LHRTTTWARR [0.010 4 ACCSGOPA 01 19-, 1 TCAqTPA Pm i- 0.01 14 RATTaT 0.010! 135 CCCFGPA 000 _128 [ADGPSNPL_ 001 98- VYCAPVP) PD 001 62 .TLGWVPQAS. 0 1.
187 LSVSVV 0.010 39 SLHSACCSG 0 010 70 _SVPLLTHPA f1 79 PSRGQALRR .008 ,F66 vPQAS v-PLLE7 N 0 1Fi241 VDPPADGS 0.005O ,f7 FHGPAFSTL I0.005 I125,. DPPADGPSN! I[-.905 F227 WF~ GPmp~CV F 005., i 6HPAQWEPVL [os 7 -TPTCATPAG 0.005 T12 SRTPTRQlj 0.005 I {V LTHPAQ 0.005 6- 1o 7iEPHPYD-L- 0.005 1 -5 2 RHLFP QEAIF J0.005, 1~ -QIPTL-GVVIPQ 70.005 178 APSRGQALR If0.005 2 7-_1--MPCSRLPPS D 0.005 f155 F-QEAFP J0.0 61PTLGVVPqA 0.005 12fTRAVTPTC 0.003 [18fPQEAFPAHP A 0.003 1 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEVIII-V4-l-LA-A1-9MERS- IEach peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight._ ISART1 SUBSEQUENCE~ S1 COR I L24i AGMPCSRL 110.J03] 138 CFGPAFST H03__ 140 l H01_GPAFSTLN__][ 03-] I TABLEVIV119-HL11A-A1 -9MERS
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
ST~SUBSEQUECESCORE!
1T 1 RLLPSLRCS 0.025 66 FJJFIGPMPCSRLL 0.025 IS I SRLLPSLR PT01 3 MPCS-RLLPS 0,013i~ 6 jSIRLL-PSLIRC 0[.0037 9 LPSL.R FCSLH -jOAJO03 2 !PMPcS RLlPi 0.600 o I CSRLLPSL[0.0F00 TAB LE VIII-V20-HLA-A1 -9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position pl1us eight.
ISTARTIK [SB SEQUENCE CORE I GDP7ASS I_ 2 fCGDPASSR 0.1507 CCS3DPASS 902 jI 5 Ei A -SRLWG 0.013- I 8 I S SRLWGAPL Duo 11 [TABLEVIII V20-HLA-A1-9MERS-
PSCA
KEach peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start L position pl ,us eigh t.
FpST!ART SUBSEQU ENCEI SCORE' PASSRLWGA 0.O 001 7, 1-9 1 RLWGAPLq 0,F.ooiJ f7 fGDPASSRLWJ 10.001 [7 S SRLGA P .000 {TABLEVIIINV21HLA1.9MERS-.
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 91 amino acids, and the end position for each peptide is the start ____position plus eight.
'ISTART SU B SEQU EN CED SCO RE 7~ [LTDPAQWEP j 1*.0 2 [AVLLTDP IF 0.015- 3 SVPLLTDPA 0.010 JYJD FPAqWEPVL tf 9.005 f ~V QSVPLLTD j .00 17 ~T [PLTDPA~Q if 0.2!5 iF-5 [LLTDPQW .0 LLTDP AQ WE 0.001i K i LPAQWEPV !fo o TBEVIIV21&22--HLA-A1= Each peptide is a portion of SEQ ID~ NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start position plu eight.
START;, ENC RE6~ I. ,1 SVPLLDL .150 6 LTDLAQWEP .125 FCl DLAQKEPVL_ 0.020 3[ 1 VPLLTDLA "o.o TABLEXIIII-V21&22-HLA-Al- 9NMERS-PSCA.- Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start positioni plus eight.J [START SU QUENO E SCORE 7W] FLLTDLAQW 10,002 [7 A LLTDLA QWE_ 0 001 JYABLEV1114v22--HLA-AI-91VERS-I
PSCA
IEach peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 1amino acids, and the end position 1 for each peptide is the start position plu s eight. I TA RT SUSEUENCE ISCOREBI 7Y7 HLAQWEPVL ,0.020 7 LAQWEPVLV 0.020 VPLLTHLAQ 0.013 2 SPLTHLA Q0O 6: C LTHLAQWEP j ,0_ I4 jPLLTHLAQW IF000 5 LLTHI.AQWE-1 0.001 IF THLAQWEPV 0.991 TABLEVIII-V24--HLA-A1-9MERSiEach peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amrino acids, and the end position Ifor each peptide Is the start position plus eight. FSTART: FSUBSEQUENCE jSCORE I I__SLTMYCT 10.030 j _LTMYVCTPV 0.025 71 VCTPPHPD [0010 8 TPVPHPDPi005 6 ~YVCTPVPHP_002 4 11 TMYCTPVP 0.00b11 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEVIII-V24--HLA-AI-9MERS- SC A.
Each peptide is a portion of SEQ ID I NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position !Lplus eight.
[START JSUBSEQUENCEJSCRP 2 SC,§ _LTMyVCTP_ =6661-i TABLE Vlll-25-HLA-AI -9MERS- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start -Position plus eight.
27 RTPTRQISS§ 0-.125- F SS i:TDP 0.030 [I ISRTPTRQIS [0.00 1 5 4 TRISSIDT 0.003 71FIs-sIDTD _PP 0.002 QISSIDTDP 000 F 7L.TPTIRQISI 0.0 6 _RQISSIDTD ]j Q.00po ;FL JPTR1S SID :0.OO TABLEVIII-V25&26-H LA-Al- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9) amino acids, and the end position for each peptide is the start position plus eight.
[SATSUBSQUECE ORE6 7 SSSDTDPPA 0030 [6{IS SS DPP ,~o 3 4 TFjRQISSSDT .10. 00 o1 L. t. ILTPTRQSS 70f0qJ L4. RQISSSDLTD 10.000 F[24 PTRQISSSD_ [0.000 TABLEVIII V28-HLA-AI -9MERS-1
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eigh [START" SUBSEQUENCEI SCOREl [9 [_SDTDPPA ',10.001l1 QIGSSDTDP 191 3= E TRQIGSSD j[0.001 117TPTRQIGSS [0.000I I=7 RQIGSSDTD 0.00 1 [TABLEVIII-V27-HLA-Al-9MERS-1 Ii 9PSA Each peptide is a portion of SEQ1 ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight TART [§UBSEQUENCE4' CORE 2 0.00F 1 Sj -RGQALRRA 0.0011 [7KLEIX-V1 -HLA-AI-IOMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10; amino acids, and the end position for each peptide is the start position plus nine.
88 F5TDLCNASGA DDSQDYYV 1.0 fI5-5 VGLLTVISK 1.000 08 ALLPALGLLL 0o.5001 _11 7KLQPAAILA 0.500_1 1-4- i 1 jW-ALGTA1 lI 0,50 F 86- CDTDLCNAS 0011 rTABLEIX-V-HLA-AI-1
OMERS-I
M PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position Ifor each peptide is the start position: plus nine.
ISTART JSUBSEQUENCE SCRE ~18[TLCSC 000 [_447[LGECWTARI 0q,450611 73[ DSQDYYVGKK jF0300 68 ,[jLNCVYDSQDPy JEQ0125 43 LGEQCWrARj .1 00 1 29IQVS.NEDCLQV 0.050 3 VLLMG 1 0.050 103 ]AAIALLP 0.o 19 LLPALGLLLW 0.050 ij-I~ LALLMAGLAL [165C 2 AVLLALLMA 0.050 10 ILALLPALGL 71 VDDSQDYYVG 005 .[TiCTQLGEQCWT 0.2 32NEDCLQVENC 0.025 I 9 LTVlSKGCSLj 477 VLLALLMAGJIO.2 72 [DdSQDYVGk 10.0201 4 4fAAILALLPAL II0.020 421 -ILLCYSCKAQV j 0.020~ [7 GHAQPAAA I_.2 i O5iAILALLPALG 40.020 :L PiLCNASGAHA 13 fALQPGTALL7020 HLQFAAAIL 0.020, 1:7 ALLMAGLALQ FO. 0 94-11[ASGAHALQFA WO 2005/014780 WO 205/04780PCT/1§S2004/017231
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus START1 SUBSEQU ENCE ISCO-RE 66 11CSLNCVDIJSQ 10.016~ IT- qt=4qi PLLLW 0.013 F875F i CCDTD-LCA 0'.010 2J L CYSCKAQ-vs 1, o I7 7~7fLLALLMAG 0 1 00 58 LTISKGCS 1:0.010 L 1 4a KTALLCYSCKA l~io9 34 DQENT 0.010i I27 K1A Q\/SNEDCL 05.010 11 LGLLLWGPG 0.1 1107,7 LALLPALGLL1 0.010 I...6511 GCSNVIDDS .1 1F 91 FLCNASGAHAL qoio [7j77 GLLTVISKGC70.1 T83 jNITCCDTDkOA1 46 7EQCWTARIRA 0.007 I100 LQPAAAILAL 0.007 161 7LIS10.005N 17 2 .CASG.AHALQ .695.
___SGAHALQPAA 0.005
T
495 T" 52 RFkIRAVGPLLTV [0005 113W~ 1LGL LLWGPGQ 0.00 i7 4 iL n~qP 0[.006 F WT9A]RfIRAVGL 0f.005_ F8 LLMAGLALQP [0_.005_ T:17 LMAGLALQPG f O.005 ff01 QPAAAALL 0 00oos F-771 Y YVGK N ITC *f00
{TABLEIX-VI-HLA-AI-IOMERS-
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10~ amino acids, and the end position 1for each peptide is the start position plus nine.__ ISTART; SUBSEQUENCESCR 82IL KNTCCDTDL 1 [003 TA_ 0.003 i s 16 q P GT A L LC Y S 0 3 L [VGLLTVISKG 1 0,003 k 39-, [ENqTLGlEq j[ 0.0031 6 27 IKGCSLNCV 0-00-2 ilVSNEDCLQVE 0[.002j E28 AQVSNEDCLO 0.002 36 LQVENCTL 0.002 F-42 jTQLGE-QCWTA 1 0.0O02 F48 CWqyTARI RAVG 0.001 I[7 SLLNCVDSQ L01 78 YVGKKNITCC 0.001 iF -[_CDTDLCNS 0.00 1 97 M HAQPAAAI' [0.001 F23[ CYSCKAQVSNIO0 F .o TARIRAVGLL00O 102 T-1 PAAAIL AL -LP [0.001 26 L AQVSNEDC[0.1 S'TLCNASGAi [H b 3 f[iENCTQGEQ 10.001
PSCA
Eahpeptide is a portion of SEQ ID; NO:8; eaci start position is spcfethe length of peptide is10 amino acids, and the end position 1 for each petide is the start position pelus nine.
STRTI-SUBSEQUENCE SCORE! I E. P P LS 62. 5001 123I DTDPPADGSi150 46 SGPSRW 1.250 43 11 ACCSGDPASY 1.000 140 TABLEIX-V4-HLA-AI-IOMERS- P..CA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
ISTARTJ SUBSEQUENCE [SCORE~ 121 6 Ti SDDPP ADG- A.00-L IF- 146: f TLPVYLRH L F 1.00 QWE PVLVPEA 201I CATPGPP 87 EAHPNASTM I[o=050L I513 LF PQEA FPA [_o.sooi 8-57,__PEAHPNASLj040 172 YqWWSPAPSIR I03 00 135 LCCCFHGPAF 0.20_ 83 J1FVLVPEAHPNA 020 69 ASVFF PLTHPAi 0.120 31 1 RLPPSLRCSL f0.100 I 2L,,I PADGPSNPLCf00 [11.ALY.GPASYR_1 01001- IF- 174 s WPAP SRGQ 00 ~F5 AVTPTCATPA 00 1h IQEAFPHPY 06 t 2.[NI-PAGPMPCS 0050 I 51EAFPAHPIYD IF005 iF -1 W-TPTATP-A] 00~ FAPVPHPDPFM f0,060 137f CCFHGPAFST F '56 1 GAPLQPTLGV 0.050 IF -16 PAHPIYDLSQ :FOOSO- 45 fCSGDPASYR 10,030 F 14671 STLNPVLRHL_ 0F.0 '1 4-1 RTTTW ARRTS .02 .130 1[ GPSN CF_0.025i Fs166 ifIYDLQVWSV 0.25 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEIX-V4-H LA-Al-i OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position Lplus nine.
STARVI SUSEQUENCESCR 1 'WTH-R-TTTWAR i[0025 'F 22 4TAPl cRJ 0.025 F113 l_ RTQIGSI J 0.025 128 ADGPSNPLCC ~[0.025 176 L SPAPSRGQAL 910_025- 11-2 J[ SRTPTR9QGS 0025 :§WGPLQTLG 0.025 42 SCCGDPAS i .2 F-1J IVCAPVPHPDPF 40.20- 16 CCEFHGPAFS 0,.020 93 STMYVAPV 0.020 177. PAPSRGQALR 0.020-? LSVPLLTHPAQ 0.02 11 PSNPLCCCFH 0015 1F75 jVSPAPSRGQA ij0.015 36 CSLHSACCSG 0.015 166 -TvPQSV-PLLT 0.013 2 GPMPCSRLPP {.1 14'8~ NiPVLRHLFPQ 0.013 178 APSRGQALR1 0.013 37RCSLHSACCS 70.010 !10.010 I23 [PAGM-PCSRL 0.010 78 4- LVPEAHPNAS F 0.OlOJ 70 1173 SVVSPA PSR(G 0.010 [143 IAFSTLNPVLR l 0.010- PMALSRTPTR ir 0,010o TABLEIX-V4-HLA-Ai-1 OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position 1_ plus nine, I[i I RI SUBSEQUENCEJ SCORE 1 -24 JFf5PADGPSN, 0,010 L 29 11 qSR LPPSLRC jJ 0.008 [7 HPAQWEPVLV 1J 0.005 F P4 I LTMYVCAPVP OO000 180 SRGQALRRAR 0.005 139 FHPFSL j0.005 F1 RTSRAVTPT 00 60 QT SGPQ 0.005 27 MCSRLPSL 0.005 RHLFPQEF 0.005 F9 ISRLPPSL RCS -,0.005 168 [LSQVW SVVSP '10.003 120 q G1QTDPPAID .0.003 50 [AY GPIL 40.003 s f[ QTLGVVPQ J0.003 4 [DPASYRLWGA 4 .003 jl:97DG-JP',S,L6U 0.0 0 3 141 4 PAFSTLNPV 1 1760 FPAHPIYDLS -J ,0.003 574 APLQPTLGVV 0.003 T X:7 PTAPAGPM4 0.003 -47 [GDPASYRLWG 4 .03 XKABEI!X-V!'1-HLA-A10-ERS- I PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is10 amino acids, and the end position for each peptide is the start position plus nine,
.FS
T
AR jjSUBSEQUENCE
SCORE,
TABLE IX-19-HL11A-A1-1OMERS- Each peptide is a portion of SEQ IDI NO: 8; each start position is I specified, the length of peptidle is amino acids, and the end position for each peptide is the start position plus nine.
STAt SUBSEQUENCE i SCORE T1] _RILLPSLIRCSIL 9 LLPSRCSLH 0.010: L 6 if:C~sRLLPSLRC 0~o.00o8 MPCSRLLPSL 0.005 H fAGPMPCSRLL 3 000 FY1 SLLPSRS 0.001 -TA X-V20-HLA-AI-1 OMERS-
PSCA
LEach peptide is a portion of SEQ ID dNO: 8; each start position is ~specified, the length of peptide is Iamino acids, and the end position for each peptide is the start position plus nine.
STR
1 SUBSEQUENO COR
E
4 [JSGDPASSRLWI 1.250Oj 2 IC-CSG( DPASRII.1 J V 1 ACCSG9DPASS [0.020 V 3 SGDPAS SRL 0oo15 8 IASSRLWGAPL j 0003-o 9 L SSLGAPLQ [0003 f 6 IDFASSRLWGA [0.003 5 JG PAS RL [0.0031 'I 10 4SL PQ 0.000 .1 7 PSSRLWGA7PF66 f000 T[ABLEIX-V21- -HL-A1-10MERS-
PSCA
WO 2005/014780 WO 205/04780PCT/US2004!017231 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
iSTA1RT ISUBSEQUENCE! SCOREl =8KDAQEV1 12.500 ILDPAQWEPL 00 F LLT P-AQWP 0.01 9j--1 TPAQWEPVL 0.005 [2qPTPAQWE (000 1 PQASV PT 000 TABLEIX-V21 &22-HLA-AI- I OMERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is Ispecified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
ISTART1SUBSEQUENCE SOE 7 LT LAQWV 1.250- 2 1 ASVPLLTDLA 0.150 131SVPLLTDLAQ 0.5 F9~ DLAQWEPVLV[0.020~ FI1 QASVPLLTDL_ 0.010 1, 4 V VL LT D LAQW [0.005Q 6 1 LLTDLAQWEP 0.1001 Y"fTLAQWEPVL 0.001 LLTD -LAQWVE [0 000 TABLEIX V22-HLA-A1-1OMERS- V- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end jposition for each peptide is the jstart position plus nine.
STRT SBEQENEPCRE TABLEIX-V22-HLA-A1 -IOM ERS-
PSCA
1Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the stat psiton lus nine.
STAT'_UBSQUECE'SCORE
3 SVPLTHLAq[jg960 j LTHLQEPV_ 0.025 9 HLQWE P-VLV 0.020 II QAVPLLTHLj000 010 FLAQWEPVLVP .005 F 4 [P LLTLQ j 0.005 6 LLTHL-AQWEP 1 .00 tLAQWEPVL- 0.001 i[ lfLTHLAQWE 0 j_.00 TABLEIX-V24-HLA-AIOMERS- 4 LEach peptide is a portion of SEQ D NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
STA [SUBSEQUENCE
SCORE
1 1 TPVPHPDPK1.5 I1 iNASLTMYVCT 0.020 8 VCTPVPH PD _002 5 _TMYVCTPVPH IF6.916 3Y SLTMYVCTPV, 0 o10i f mii~yvcpvp 6-0005 19 IF WPPHIPDPP 0.003 2 ASLTMYVCTP f0.002 f77 VCTPVPHPD [T~ FTABLEIX-V25-HLA-A1-1OMERS- INO: 8; each start position is sepecified, the length of peptide is 10j amino acids, and the end position for each peptide is the start position plus nine.
STRSUBSEQU ENESCORE If2 ][STTQS .025 iF TPTRISS ;i0.025 1 Kio~sslroPPo .003 [71 TR- SID 0.00 8 QISITDPP 0.001 7-K7LR9ISSDTFLg 0.001 FT TRQI1S-SIDTD- 0.000 4 4 TPTRQl SSD 0.000.C ii TABLEIX-V26&26-HLA-Al Each peptide is a portion of SEQ ID~ NO: 8; each start position is specified, the length of peptide is amino acids, and the end position Ifor each peptide is the start position START, SUBSEQUENCE~ SCORE'I 9-1 I S DTDPPA 000 77 iSRTPTRQISS 0.025 -3 RTPTRQISSK 0.026 10 4 ,SSIDTDPPAD 10.003 E.7 117[ LSTTQIS 0.002 5 IPT §!SDT1 0.00 1J 8 QISSIDTDPP [0.001 1 4 qISSIDTDP 0.000 6-[TRQISSIDTD 1[000 4[TPTRQISSID 0000F I TABLEIX-26-HL11A-AI-10OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position isI specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
I9 SDTDPD 1.5001 L I RPTRQGSS .025 II7 ISSDTDPPA. .0 WO 2005/014780 WO 205/04780PCT/US2004!017231 [TABLE IX-V26-HLA-AI-1OMES'
P.SCA
rEach peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the ___start position plus nina.
SART FSUBEQENCE S CORE 8 GSDTDPAD 0.003 6j QC IGSSDDPI 0001 3f PTQIGSSDT 4 0000- 4jT]fTRQIGqSSDTD 0.000ij FfTO SDTDPPADP IFooco: TABLEIX-V27-H LA-Al-i OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
[STR UBSEQUENCE[SCOR E' j24RGQALRRAQ 0.001 LIIII[PSRGQA LRRA [ThO00: TABLEX-V1-HLA-A0201 -9MERS- [Each peptide is aportion of SEQ I D NO: 2; each start position is specified, the length of peptide is 9! Iamino acids, and the end positicn for each peptide is the start position plus eight.
43 1[ 9LGEQCWTA 162.756, [MGL 83.527, [7 F LLMAGLAL IF79.041 109 FLPALGLL f36.3161I [105I A LLPAL F. 99 108, ALLPALGLL 23,6 33 IL7 7I1LLQPGTALL 1[1'H362' FTI I L Y7 K ~'[1s.38V j--i7[ LLLWGPDGQL 117.4681 .TABLEX-Vi -HLA-A0201-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9, amino acids, and the end position for each peptide is the start position plus eight.
START SUBSEQUENCE SCOREj 42A TQLGEQCWT 115.375 LV~ ALQPAAAIL f85 d8446 YK 7 SNEDcyjf3.6 83 NITCCDDLj~ 78 YVGKKN-ITCj 2.00 60 VISKGCSL 1.869 107 LLALGL. 1.866 I 13 '1LALQPGTAL 1.866] 4 VLLALL AG [1.078 112?8 AQVSN EDCL 1.06T [15[LQPGTALLC 0.856 Fi LQPAAAILA 0.86 [jY4GALPGA 0.646, :[5[GLLTVISKJYG4 71 7DDSDYYV, 0361] 101 QPAA-TL7 0.29 47 'QCWTRIRA 0.269 A7NTI~LLL LM 2 63 fSKGCSLNCV 10.222J F81LLAGLLQ 40.216 1 E4QCWTARI 1 0.203 1i~ A5GLALQG 'Ri so AILALLPA i ~s 92__[FCNASGAHAH[ 0139 F_ 54 RAV -i iIV 1 0.37j F-jo 6 ILALLALG 0.127 27 KQVSNEDCI[j 16- 18 FT MKALLALL 0116 I'tTj0. 0 7! TABLEX-Vl-HLA-A0201-9MERS-] Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start L position plus eight.
FSTARTI SU BS EQU E NCES L ~iGTALLCS if 0.69 j96 LGA HALQPAkA1 F0.
84 ITCCDTDLC; 060571 6 LALLMAGLA 111 1[ 86 CCDqTILT LCNA 0.30 E7:VDDSQDYY 4 0.029] 9[ DLCASG ~[0.026 'F l 7--LC YSCKAQ IF qo? 1 0 ARIRAVGL I0.023 9 L-FM -AGLALP 0.018 98 _oo V6-1V -IS KGC SLN O017J I 102 1AA-AIiZL-L-' 0.015- 35~7 CLQVENCTQ 0F.0151 34 DCQECI.3 F6 7 SLN-CVDDSQ ,~0 1 8 CWTARIRAV F-004 I10 1 'AGLALQPG 0.004 97 AHALQPAAA_. 3 [667 GSNCVDDS 0.0 85 CCDfTCN [6.002 69 [_NCVDQD 002 749- WTA R IRA VG 7F-6- o] 7 F-)YGKKNIT Po'.oo [IIO-P (LG LLW :1 0-002 56 [:VGLLTV\ISK 0[.001l WO 2005/014780 WO 205/04780PCT/1§S2004/017231 [TABLEX VI-HLA-A0201-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start osition plus eight.
[START S§UBSEQUENCE SCORE 29 VSEDLQ 000 19 TA LLCYSC 0.001- 1 6 -QPGTALLqY fQ0 F-111 FPA LGLLLWG 0.001l 4j C TQLE EQCJFO001_, 907 DLCNASGAHN 0.001 I1 LGLWP 0.001 33~~E ECQ E ,0 JE FNEQCLOVEN I 0.001 87 CTPLCNAS 0.000 37 QVECTQLG 0.000 76 PYYVGKKNI _IF.0007 175 Q1[DYYVGKKN 0.000 93E FNA-GAHALq 000 TABLEX-V4-I-LA-A0201-9MERS-: _PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9i amnino acids, and the end position for each peptide is the start position plus eight..
1[66 YDLSQVWSV 28.361 146 TLNPVLRHL 9.827 I170 _QVXNSVVSPA 8.298 Y TYVCAPV 6.076 167 DLSQVWSVV 3.636 TABLEX-V4-HLA-A0201-9MERS- Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start p -osition plus eight.
START. SUBSEQUENCEF§ SC0 REI 8- .4 LVPEAHPNA 113. 00 157 APLQPTLGV I .8 1 60 f PAHPIYDL 1.475:, 80 WIE PVLYPEA 11 1.22
MTHRTT
1 TWA 11 0.45 1 5[WLPTLIIFa411- 66[VQASVPLL1IRS4SI 3F 2 LPPSLRCSL F0.23 T 58 4PLQPTLGVV I[ 0.18 1142 AFSTLNPV i.11 9211ASTMYCA 0.18 24__ AGPMPCRL 0.3 1120 4GSIDTDPPA 11 .1 1 F39[ FH GPAFsTL Lb 1 53 [RLWGAPLQP40.124 176 MALRTPT 16~ VTPTCATPA1L0.117~ F7Y GPSNPLCCC [0.075 6 Lv-vPQAS 6 0075; 21 -FATPAGPMPC 0. 69 8 WARRTSRAV 006 N1_ASTMYVC j0057 1F02!, VPHPDPPMA [0.0-55 I,90 PN, ASLTMYV [0.055 78 ,AQWEPVLVP P-0048 TABLEX-V4-HLA-A0201-9MERS-I
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is 9 specified, the length of peptidei9 amino acids, and the end position for each peptide is the start position plus eight [START [SUBSEqUENCEJSCOREI 75L [THAqWEPV 0.040 1-63 HPYLSQ-v 0.036 93 SLTMYVCAP 1 0.034 RIFPSRCS _f EX34 I14 LFP QEAFPA j, 0.034 42L7 SApCCsGDPA 0.034 .1 37 TRCSLHSACC 0.032 F138 CFHGPAF STT0.03 135 LCCCFHGPA 10.0271 49 PASYRLWG0.026 JTfRTTWARIRT 002 [155 FP QEAFPAH ifo0.17 ~7 PQASPLLT 0.017 [110 ALSRPTR (0.015 97 YVCApPPHP flo0.014 61 fPLGVVPA If0,013 153 H1LFPQEAFP ]l70.01 [IFT3 CCFHGPAFS 0. o di 101_ PVPHPDPPM 0 .0io0 6 [PEAHPNASL ;f0.009 1[4f STLNPVLRH 0.009 IN PPMALSRT 0.008 7 f EAH-PNASLT 0.00 8 128 ADGPSNPLC Fa000 f~7fPASRGqAL ]o [jf4i3, AFS LNPVL ]f om iF 72T F PTHPW][a06 [F 1517 LRHLFPQEA ~F ooo -17Ef SPAPSRGQ 4'qq L0.004 :jigj 7TCAT-PAGP 4 000 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEX-V4-HLA-A0201-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9! amino acids, end the end position for each peptidle is the start position plus eight.
[S-TA RT SUSEUECSORE.
LWGAPQPTl C604- 1j3Isoi V LHFPQE i[ 00 169 1[ QVWSVVYSP 1[0.003 A.H PNASLTM [~o 2=8 PFCSRLPPSL] 0.003 132 J[ SN PLqCCH. ,[0.00 L[74 FT HPAQWEP 0.003 127 1[ ADGPSNPL '110.0-03 RLPPSLRC -1[0.003 [129i DGSNLC 10002 ASVPLLTH 10.002 172[ WSVVSPAPS 0.002* 4 'CSGDPASYR 0..902, L18 QIGSIDTDP_ 0.2 F76 HPAQWEPVL 0.002 27 1'MCRPPS~ 0.002 -31'CSLHSACCS .0 [IIIj LSRTPTQ [.0 6 1 TTWARRTSRk F[ .00 103 PH FD A 0.001 7173 j SVVSPAPSR 0.00 [TBLEX-VI 9-HLA-A0201 -9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9', amino acids, and the end position for each peptide is the start position plus eight.
[S-TART, SUBEQUENCE SCOR E 7Y7'LLSLRSL 36.316: LLPSRCS 0. 127 ID LR- GPCSRLL [0.10 }rTABLEX VI 9-H llA-A020 1-9MVERS- Eah ,PSCA Eahpeptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptidle is the start IL position plus eight.
![STARTI SUBSE QUNCEIIS:OR E 4PCSRLLPSL~j [E107 6 RLLP SLRC 00[ 3 MFPCqSRLLPSI0.002j 2 C PMPCSRLLP 0.000L I:TABLEX-V20-HLA-A201 -9MERS- PSCA Eahppieis a portion of SEQ ID NO. 8, each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start position plus eight.
TRT USEQUENCEI SORE 3F GPSR 0.056 6- [7hA SSRLWGA 0. 0 2 6 8- c SS LA 0. 011 2 CGDASR 0.000 V- [DT-3S"R L 0.000' 4 GDPASSRLW [0.000 9 SRLWGAPLQ 0O.000 ITABLEX-V21 -HLA-A0201-9MVERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptidle is the start l position plus eight.
START SUBSQUNC COR E 6 LTDAQWE I 0.571 3 VPLLTDA 0.213 F DPAQWEPV j-0. 080 1 ITABLEX-V21-HLA-A0201-gMERS-
_P.S.CA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 Iamino acids, and the end position for each peptidle is the start position plus eight.
[-TART s~u-PsEQUE NCE~ ISCORE t LILUDPAQW ~I[000 PAQWEPVL .0 IF7 LDPAQWEP U[ oqooi QFAS11 VPLLT D 0.000,Q 4L 2 7[ASVPLD =0.000 .F TABLEV21 &22--HLA-A0201 9MERS-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptidle is the start position plus eight.
START S-UBSEQUENCE' SCORE 8 R DLQWPVL 065 ]LLTDLAQWYE 0.571 (77 TDLAQWYEPV i[ 0.298 2 SPLLTDL\VFi[021 SVPLLTDLAQ '[.001 fTABLEX-V22--H LA-A0201 Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9, amino acids, and the end position for each paptide is the start position plus eight.
[si-AR' SUSQEC~SORE 12 FsVPL LTHLA F[ 0,436 17 117' ASPLHL [.2 F 8 'HLAQWEPv-L [0.I298 WO 2005/014780 WO 205/04780PCT/US2004!017231 TABLEX-V22--HLA-A0201- I I 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9' amino acids, and the end position for each peptide is the start position plus eight____ 7 THLQWEV 049_ PF LLTHLAQWE 0 139 4 LLTH[AQW j F671 LTHLAQWEP 0003: Y[ PLLTHLA 0.0011 STABLEX-V24--HLA-A201- I F 9MERS-PSCA iEach peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 91 amino acids, and the end positionI for each peptide is the start position plus eight.
,FOiAR[ [BSEQUENCE jSCORE' ff ~LTMYVCTPV j 6.076 i [F AsLTmyvcT .27T 7 2 SLTMYVCTPF53ia 4 TMYvCTPVP 0.0914, 6 YVCTPVPHP 11.014 IiE~ P HPP- q 0.000 I]E[MYVCTPVIPH [OA0jO TABLEX-V25--HLA-AO201- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positionI for each peptide is the start position plus eight.
STARTIPO'BSEQUENCEi[ SCOREl 3 ITPTRQISSI 0.167 L 9 SIDTPPA_ .1 7 QSID TDP ~FF 0.02 1KTTiTRQISDT 0001 6To RTABLEX-V25--HLA-A0201- 9MERS-FSCA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start positio plus eight. SSTRTJUBSEQUENCE, SCREI 2 j RTPTRQISS 0.001 T IFRo If 4 PTRQISi 0o.000] [A EX-V25&26--LAA02D1- Each peptide is a portion of SEQ 2ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
START ISUBSEQUENCE SCORE 1 7 SSDTDPA i0.133 f-7 5 QISSSDTDP jO. 0.002F F17 FTP TR QIS 0. O1 7Y7 RQISSSDTj 0.0,01-1.
LSSS§DTDPP 1 .000 2 fPfRQISSSD j0.000 TABLEX-V26--HLA-A0201- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start Position plus eight.
S TA RT,, S U BSE Q U E N C E, fSC El I7 GSSDTDPPA 0.1331 FQlGSSDVDP 0.002 if 7[TRQGSDT 10.00 1j 4f77 RQIGSSDTD I0.001 F 77SDTDPPADG-!F -6 0.000 T 1= [TTRQ(IGS 000 :6S-D-TDPP IF 000 ITABLEX-V26--HLA-AD201- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
f§TL SUBSEQUENCE! SCO6RE 8 ESDTDPPADi0.0 2 PTRQIGSSD- 0.000 If BLEXX2T7HLA-AO201OMES Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position f or each peptide is the start position _,plus eight. STT SBSQUNCJ[-SCOREI If- _7 RGQALRRAQ 0r.0001 ATABLEXI-VI -HLA-A0201 Each peptide is a portion of SE ID NO: 2; each start position is specified, the length of peptide isI 10 amino acids, and the end position for each peptide is the j start position plus nine.
IFSTART LUSQUNE SoRE f7-iE FALLPALGLLLJ79041 F701 CD QDY [54.8941 106 ILAAGLF F36.316 f 7 jdLQE-NCTQL. ,21.362; 57 ~L T7S 18F2.382 1 7i7 TQ2LGEQCWTAf13.978 FiT 7[ GLLL-WGPGQL. I f7-o7fLQP A AALZOEi 8.t-459 1 99 [ALQPAAILA f4.96881 7W4 LQPGqTALlC f 4.9688 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 fTABLEXI-VI-HLA-A0201- Each peptide is a portion of SEQ1 ID NO: 2; each start position is specified, the length of peptide is amino aoids, and the end position for each peptide is the __start position plus nine.
[TAJYVKN6ITC[.599 1 [f13:fLALQPGTALL if866 47j'qCWTARIRAV N.33 {52 If RIRAVGjLL 1.672z [49 WTA-RIRAVGLl i 13T61 F611lf VISKGCSLNC ,1.t _1-04j AAiI-LALLPAL Ef 0.682-1 i41 fCTQLGEQcwT1 Fjo.*9 V'i07 LLLPALGLL1[.51 KAVLLALLMA 0.555_ F:T27 KAQVSNEDCL]f[509 59111 LTVISKGCSL q- 0.504- 82 KNTCDL 0.488 fo DLCNASGAHA[0.7 37NITCCDTDLC TLQ33 85 -FTCODTDL.CNA 17. O6 f109 20. LLAGLWi 91 f7.~ TALCYSCKA '1o0256 :91 jLCNASGAHAL 1 0.27~ 9 IF' 1 LMAGL-ALQG 'f0.210 [MAG LAL QPG .7 AAAlLALLPA [0159 7 7 AL -MA GLA LQ 0. 12 7 f- 77 LLAGLQP 1 0.094,1 gf 1['7A- GA LQ PA 10 0.075 f2 96 GARHLQP 1[ 06gi1 If 62 F ISKGCSLNCV if 0.0621 F77AVLLALLMAG .5 f -QYYVKKN 1' 046 [TABLEXI-Vl-HLA-A201- 1OMERS-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino aoids, and the end position for each peptide is the start position plus nine.
TARf-T SUBSEQUENESOR ER 312 JNEDCLQVENC 0.044] 905 fSA-1LLLPTiFO03 1FYA~QPTA~FO -T -1 MKAiVLLAL .012E q '1 102 LAG-LLLWG 001 563 VLLTVISKG 50 TAR RGL L 000 60 T VIGCSLNO.077 F16 QRPGTALLCYS ]0.006 28 CKAQVSNEDC'7fl 03 TABLEXI-Vi -HLA-A0201 10MERS-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptidle is i 10 amino acids, and the end position for each peptidle is the _start position plus nine, STA:RT ISLUBSEQ-UENC;E SCf 113 [LGLLLWGPGQ' 0,001 j GCSLNCVDDS $0.001 40 NTLEC~~7 54 RAVGLLTVIS j~000 87 CDTDLCNASG 64 KGCS LNCVDD F 0.000j 18FALLCYSCK LO.oOq:] 111 PALGLL WGr, 0.000J 93i NASGAALP 0.0 86 CCTDLCNA 00 66T FC SLNC(VDDSQ, F.007L 38 EvEN C-TQLGEq- 0.00 31 [SNEDCLQVEN F ,000- 76 [DYYVGKKNIT 0.000D TABLEXIkV44'LA.A0201- Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptide is the start position plus nine.
STAR-T SUBSEQUENCE'
COE
L53 .LLGAPLOPT 14.59 IF 31 [._PSLRcsLfi121.3621 83 71-- VLVPEAHPFNA 8,446 I6i 651 VPQSVPLIT 151HFQEAA 3.2 :7 [VRHLF- 1PQEAII 1.39 1 LSRTP-TRQI I .0871 6 GVVPQASVL, 1.69] 745- LTHPAQWEPV 1.368]1 1777 55 7.7f8 YYVGKKN!TC.~ 0.003 SQDYYVGIKN f'1 0.003- V-GLLTVISK 0O.00-3] IRA VG LTVI1 07.0029PT DTDLCNASGA f0.002 ITCCDTDLCN f0.002 AQVSNEDCLQ' 0.002i 'f 39 -[ENCTQLGEQC if0.00 SfLYSCKAQVS f oI 24 -jc~Q~fo01 I ~~4LNCVDSQD1L.001 2 7 fGTALLYS~j[0,001 Fj 7YSNEDLQVE1[ 0,001- WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXI-V4-HLA-A0201- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the ;L start position plus nine.
SUSEUENCEI SCORE [16-9 S S S [159- ~1 1{SQVWSW1[:A: [141T F G-PAFS TLNPV iF 1.044: F37 CFHGPAFST[1.4 6 [TWRRTSR I0.573- 'CSGDP ASYRI.7 1 ASRLWAPL [.4 H5JfROSLSAO~0.37 iAPLQPTLGVV '_0.20 {7 LLTHPAQWEP 0.190 TY,CAPVPI 0-17- LI6 SPAPSRGQAL [0.139 165 IYLSQVSV 0.113 162 ifAPYLQ 0.111 91 N _ASLTMYVCA 0. 1041 87F 1 -0.08 [7K IY_ 1_PQASVPLLT[ 0.0F8317 [bPTLGVVPOA 0o~75 14673TLNPVLRHLF F.075 I ATPAGPMPC I-6- 11 RTSRAVTPTCF 0.06 119 -IsIfDDPPA 0.055 134_1PL6CCCFHGPA 04 84_, P LVE NAS I 45 F-i3, TPTRQGS 0437 17K81 D!PASYRLWGA P0.042 1, I[~~IAFPAP D I 0.0 693AvPLLTHPI 003 ITABLEX1/4-HLA=A0201-.
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
START SUSQEC SCORE3 F138 CFGASL 0.0281 i76:PIYDLSQVWS 0.63 7=1i HPAQWEPL 0.015 8 T -WARRTSRAVT, 0.015 126 PPADGP(SNPfL 01 170 _QVWSVSAPI 0.011I 142 PA FSTLNV 0.1 1 O],jPVPPPPMA]0.0 75 VS PPSRGQA1[0.0071] I7_5&J FP79EAF PAHP .00q7 i~ ADGPSPLCC I[0.=007j :72 [PLLTHPAQWE 0o.00J 23 PAGPMPCSRL' __0=0 1f73 1 3f 97 ?[YVqAiPVlPHPD. f 000 ,[129 T71DGPSNPLCCC i[ .0061 [87 [1,iHPNASLTM1L0.005-- 121F 7IDTDPPADG §6.004 [7 3PPMA SRTPTi004 88 F PEAHPNASjLo .0 1D32ESNPLCCCFHg1 0.003 26 PPCSRPPS 0.003 I 36 CCCFHGPAFSjf 0.003 f i MTHRTTTWAR 0.3 1__29 CSRLPPSLRC, 0.00 [TABLEXI-V4-HLA-A0201 1OMERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start Position is specified, the length of peptide is 10 amino acids, and the end jjposition for each peptide is the start position plus nine.
STARTU BSEUENESOR 10 RRTS RAVTT [:02_ 21, AiT PGPMCS 0002 167 DSVSV .0 38: C.1SLHSACS§ .0 16 VTTCTPAG IF.99L2 16 PQEAFPAHP 0.00 109 Y MALSRTPTRQ 0.001q [1 2 3 SACSGPA .00 i 14 7L CLRVLRHL Fi- 0.001 139 FHGPFTL 610 TABLEXI-Vi 9-H LA-A0201-
IMERS-PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the 0 start position- plus, nine.
:sT RrSUBSEQUENCE SCORE _8 FRLLPSLRCSL 790411I 4-9 SILPSLR CSLH 7 q AGPPCRL I 09.028 2 1 GPIOPCSRLLP WO 2005/014780 WO 205/04780PCT/US2004!017231 -TABLEXI-V19-H LA-A0201 1 OMERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the L start position plus nine.
STARTJSUBSEQUENCE
SCOR
7 RLLPSLRCS 0.0006 PCSRLLPSLR 0.900iT-ABLEXI-V20-HLA-A0201- Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position L. plus nine.- [STARf SUBSEQUENCE:
SCCRE
3 CSGD PASSR 0.572 F87 A'SSLiIAPL 0.139 6 [DPASSLWA 1 0.042 f GDPA:.SRLWGI :F-0.1 1 1_fACCSGDPASS .0 2 CCSGDPASSR) 0,000) 4iED.,SGDPASSRLW f0.000 _kjLSRWAPLQ_1[.000 7,11[ASSRLWGAP 0.0 FTABLEYJ-V21-HLA A0201 -1 10MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptidle is the sta! psition plus nine., [STAI Ti JSUBSEQU ENCE,, S CORE.
SLLTDAQ 1EP F0.7779 8 lT7[ TDP AqWEPV 'L0 4.
F fASVPLLTPA f0.016 F 1TDPAQWEPVL I0012 j~7fPLLDPAQE f0.007 TABLEXI-V21-HLA-A0201- 10MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
4 SVPLFOA *j00 1 2 QAVPL LTP T-1 00900 1 l TABLEXI-V21&22-HLA-A0201- _.....1OM1VERS-PSCA Each peptide is a portion of SEQ III NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
JSTRT UBSQUECE;SCOREI 1'L9! [DLAQW-EPVLV ii 5.216 I 1 QASPLLTDL 06782 TID .QWEPV 1 0.547 2ASVIPLLTDILA ii 9.016 LLLDLAQW -0,.007- 5 FPLLTDLAQWE 799 :7D7 SPLPLTDLA f .017 TABLEXl-V22-HLA-AO2Ol- 1OMERS-PSCA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
STA [SUBSEQUENCEi[SCORE; F97 HLAQWEPVLV 12.365 f77 LTHLAQWEPV 1.368 1 _QASVPLLTL 08 6 jLLTHLAQWEP [Y ASVPLLTHLA IF 0.932 STABLEXI-V22-HLAkAO2OI. I Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 101i amino acids, and the end position for each peptide is the start position1 L- plus nine.
STAR [SUBSEQUENC SCORE1 -1 4 PLHAW .0 II:7! LLTJHLAQWE 0.0 SVFLLTHL Q 0.001 L QWEPV-LVPE =000 FTABLEXI-V24-HLA-A0201-1OMERS-I PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for eachI peptide is the start position plus nine.
.FsiA, [SUBSEQUENCE I SCORE) 11 J SLTMYTCTP V 69.5521 J 5 fTMYVCTPVPH 0o172 V ~N~SLTMYVCT 016 10 fTPV PHPDP-PM 0.032 f 7 -YVCTPVHD1jj 2 LTMIF VCTHiPP Fo.oo 6 MY[h VCTPVPHP 10.000 TBE-V25-HLA-A0201 -1 OMERS- TABLXI- PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
fSTART SUBSEQUENCE JSCORE 7 RQISSIDTDP 0.2 Q SS I7 DP P F6-6 0017 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXI-V25-HLA-A201 I OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for eachI peptide is the start position plus nine, I!T fSUBSEQUENCE SCORE! SSIDTDPPAD [0.0001 [4 TPTQISSD ~0000 ,F LSRTPTRQIS 4F0 000F 4 2 f SjRTPTRQISSJ IL9P 6 IRISDD 0..000 TALEX-V2E&26--HLA-A0201- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end posidion for each peptide is the start position plus nine.
[ST-AR SUBSQE SCORE 1 D ISSSTDPP 0.055 ~j RQISSDTP][0.002 7Y[STDP7AR ]7 iLT ass T [I o9-t9 77TRQISSSDTID FO 0 _0 TABLEX-V26--HEA6A201-1OMERS- Each peptide is a portion of SEQ ID NO: 8; each start position is specified,' the length of peptide is 10 amino acids, and the end position for eachI jpeptide is the start position plus nine.
I START S~UBSEQUENCE SCOREi Ifl GSSDTDPPA 1 0 0.5 .5 [RJQSSD DP 0.002 f 6 4QIGSSDTDPP 000 F q STP PA 0000 j ,o 6 If T7 If0 RTTQS 0f 0o1, F gT71 SSDTDPPADG 110 000q, [7T PTRQIGSSDT 0,000 j[io SDTD PPADGP 0.0 E774 TRQIG(ssDTDf0.000 TABLEX-V27-HLA-A0201- 10IMERS-PSCA___ Each peptide is a portion of SEQ ID NO: 8; each start position isI specified, the length of peptide is I10 amino acids, and the end position for each peptide is the L start position plu nIne STARTI SUSQECE SE 2 SRGARRA 000 TABLEXII-Vi -HLA-A3-9MERS-
PSCA
Each peptidle is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position, plus eight. _1 FSTAR UBSEQUENCE
ISCORE.
LJALMAGLL 11.800 iO 9 1 LLPALGLLL 1.200 LLAMGl- 0.900 99f LQPMI ii 0.900 14j ALQPGTALL 0.9006 4 jfQLGFQCWTA 0.900 1 20 LLCYSK 10.0 4108 1 LPLL !I 7T 11 ITALLCYSCK 0.3006~ 115 L[LLWGPGQL 0.70W 14 :1 LLLWGPGQ 10.270 [5 GLLLTVISKG Io2~ 5 tVGLLTVISK 10.18 0 F V GLAL QPGTA7f F0.180 4 5 4 LTvIcsKGC F-1__ 4105 4 AILALLAL .135 fTABLEXII-V1-HLAAMMERS-I
PSCA
IEach peptide is a portion of SEQ ID INO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
JSTAR [SUBSEQUENCE
[SCORE
1[-711QPGTALCY if 0-120 q _11 A LGL L G_ _fo~ VLLALLMAG [090] f 7fGTALLCYSC f09 if: E3]if DSQDYVGK IF -0.09 IL6 NCVDDSQDY f06 fLNAGLALQP f0.060 f8 fLLMAGLALQ f0,045 f'7fj LQVENCTQL f04 ,F YVGKKNITC [0,040 4 67 SLNCVDDSQ f0.030 Thi' LALLPALGL 40.027 -2-8 if AQVSNEDC-L -0.02 7i 4 44RAVGLLTVI[ 0.020 iF1CLQVENCTQ 4 00-E20 106-, -f ILALLPALG f0.020 jfi'To" zQP-AAAILAL_ 0018 F7 15 FLQPGTALLC 0 01-8 2 f KAVLLALLM [0,018 F 90 DLCNASGAH '4018 F45 GEQCWTARI F016 13fLA QPGTAL f0.013 f 9 f ALPAAAI 0.0134 ffoio I LQPAALuA f 001 41fCTQLGEQCV JF 0010 IT-CCDTDL-Cif01 f7fLLCYSCKAQIf01 LCYSCKAQV F0.010 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXII-VI -HLA-A3-9MERS- C..A Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9~ amino acids, and the end position for each peptide is the start position I[ plus eight.
[SA!SUBSEQUENCE
SCORE
4211 TQLGEQCWT I[f 007' jL-PALGLW ]f0.006 96GAHALQPAA -f[_.006 01 TARIRAVGL 0.006 IAV-GLL-TVIS0.4 [44- LG EQ WTAR.- f 0.004 ISKGCSLNC J0.003I NTQLEQC 0.002_ I 7 QVENCTQLG 0.2 61 \SKGCSLN 0.2 91 LNASGAHJA 0.002 29 QVSNEDCLQ 0.0021 I4 F W TARIRAVG I 0.002 10 PAAAILA LL 0.001 [921 CNAsGAAL J .m001 15-9 LTV1SKGCS 'FOOT 166 _SNVD .0 -6 FLALLMVAGLA 0.001 51K ARiRAVGLL 0 .001 DCLQVE NCT ~o 71, VDDSQD1(yY f .001 F7 7.IF VGLIT 001f~ f7IGCSLNCVDD 0 .001 D6 ~jDYYVGKKNI .000 I1 AAAILALLP 0o.o0o TCCDTDLCN 0f .000 IF 7 NASGAHALQ if0.000 TABLEXII-VI -HLA-A3-9MERS- I PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position pllus eight.____ f~SUBSEQUENCE SCORE __81 KKNITCCfo0 [LiESG CSC[ 0.0 L7 fDkLNASGi1 0.000 f ifSGAHALQPA IF 0 0-00 _f94 _fASGAALQP kfooo- E.33 f EDCLQVENC .i 0.000 f11~ AGLALQPGT if 0.000 111 ]..._PALGLLWG 0.000 38I VENCTQLGE if 0.000
FSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight._ 1,1TART I SUBSEQUENCEIj SCORE 7fTTVVARRTSR T-1-666 F N7 yMSVWS SPA IOASO45 f35 SLRCSLHSA .1-6300 146.[iNVRH L f0.20.3 93 S. 0.180 f s fHLFPQEAFPf1,[o.T5 F145 ,fSTLNPV-LRH 0.135 Do "ms vc f TMYVC FPVo_ .100 158 f EA-FPAH-PIY 0.090g 167 I DL s QTvWSV 0.
TABLEXII-V4-HLA-A3-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight. START 1SUBSEQUENCEI SCORE! 15 0.060 Ei F5 PASLW, 0.060 S TLGPQAS 0.060 j109 MAl-SRTPTR_ 060 [7837 _VLVPEAHPN 004 I 16 IFAHPYDL 0.041 F 7[AP SRGQALR 0040-5 PL -THPAQ1W 0. 0 CSGDPASyR 003 73 [LTPAQWE 03 KI FLTMWyVAPV 0022 144 FSTN 0LR .o020 739 ISLHSAGSG_. 020-1 70 SVPLLTHPA 020
IIF
7 FT GMC 0.020 F136 CCCFHGPAF 0,.020i 66FVPQASVPLL 0.018 76 LPAWPVLf0.1 11 TSRAVTPT 0.0151 64e7 G/VPQASVPF0.013 F139 1 GPSNPLCCC0.1 TT TWA 0,0 ['.12 C 617~ PL-GVVP~QA 1 0.010 108rPMALRTPT 0.010 NA-SLTMYV C I0.009q- WO 2005/014780 WO 205/04780PCT/1§S2004/017231 FTABLEXII-V4-HLA-AM-MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight___ [START SUBSEQUENCEf SCORE L156 1 FPQEAFPAH 0.009 F-577 APLQPTL-GV 009 F66 QAVPLT F.09 31 LRLPPSLRCS_![j0.009 23 4 PALGPMPCR jO0.0061 179 4 PSRGQARR1400051 1342 I PL HP 40.0065 '1 152 f N RHLEF .0-05 H 120 GSIDTDPP 0.005 17 16 PIDLCSQI 0.00 LTHPQWP 000 [17Lf RIITP _G 1 50.004 12 ff PMPSRP 0.003 PVHDM 1 0.0031 174 j WSPPSRG 0.003 f169 jfQI SP 0o.o3 1i 1 jf VPF- P MA 0.003 -T7_ If qFPASTL 11 0.003 F-K47f RAVTPTCAT [0.002 I 11f RSIDTDA ]FO 002 4[ 2 -fSACOSGDPA f0002 f13 IfCCFHGPAFS 0? 002 I 1 f YLWAPL if0.002 TABLEX1-V4-H1LA-A3-MERS-
PSCA
IEach peptide is a portion of SEQ I D INO: 8; each start position is specified, the length of peptide is 91 Iamino acids, and the end position Ifor each peptide is the start position eight.
STARTI[SUBSEQUENCEI SCORE! ,I 135 LCCHP f0.002 80 WEPVLVPEA f -0.002 F13-1 1 RPSNPLCCCF fl 0.002 1 4 RTTTWARRT 1f10.002 E PiL AF S T LN PV 792,1 ASLTMYVC 0 .002 1D 2 FTSRAVT pC If .002 22-E [ITPAGPMPS 0f.001 ff10 4HPPPMAS jf0.001 If 30 4 SL PSLRC-- 0f .0o1 14fPVLRHLFPQ 1f 0.001 127 PADGP§NELl 'f0.001, 81_ fEPVLV EAH if0.001 14-1 I GPAFS§TLN [6.60 105LPPPALSR1 0.001 F49 FASRLGA40001 5_fWGAPLQPTL 40.001 f 24 L[:PG4PG.PCRL 40:00! TABLEXII-Vi 9-A3-9MERS-PSCA IEach peptide is a portion of SEQ ID: NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start position STAR SUBSEQUENCE I SCORE 8 hLLPSLRCSL4 0.6001 CSRLLPSLR 0 .020 [7 RLLPJSLRCS ~[0.013 [1 GPMPCSRLL if0.004 1 2 _PMCRLLP If0.004 9 [T LPmSLSHP _.0 6'L SRLLPSLRC .011 TABLEXIW-20-HLA-A3-MERS-
PSCA
Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
fSTART SUBSEQUENCE SCR If 8 IF SS-RLWGAPL 0f 009 6 ASRLWGA [f 0001 F-7, cSGPDPAS fI 000-6 1- 4 IPPASSRLWG "1[0000 I4 4 GDPASSRLW 10.000 If~ fASSRLWGAP f0 0.
TABLEXII-V21 -HLA-A3-MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight. TSUBSEQUENC.E SCORE! 5PLLTDPAQW 0.030 SVPLLTDPA 0.020 97 7 PAQWEPVL",j[0.,005_ E-7 ilLTDPAQWE .0 'fT [Th--LTD jf ooi' 27 ASVPLLTDP 15 0.000 4T~ VPLLTDPAQ IF0.0 8 i TDPAQWEPV [00 TABLEXII-V21 &22-HLA-A3-I 9MERS-PSCA__ j Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide i s 91 amino acids, and the end position for each peptide is the start position plus eight.
WO 2005/014780 WO 205/04780PCT/1§S2004/017231 IT7RR SUBSEQUENCE:
SCORE:
F[8_JID L)QWEVL ,0.540 [4 1 PLTDLAQW j0.020 0.020, [ill SVPLLTDL r0,010 63 1 LTDQE iL 99 =[ll TLAWEP i[0 [TABLEXI-V22-HLA-A3-9MERS-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
8 LLAQWEPVL 3[ 1.800 4 31 PLLTHLAQW 0o.0451 l 5 FLLTHLAQW I 0.90 2_ iSVPLLTHL__ 0.010 I 6j THLAQEP [.0 L 9 [LAQWEPVLV._I 1F1, Y LLTHLAQ 4 -oo VF7 L THA IQWEPv 0.00066 TABLEXII-V24-HLA-A3-9MERS-
PSCA
Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start ____position plus eigt I777[SLTMYCTP 0.100 3 4 LTMYVCTPV [0.2, J64 ,YVCTfpvPH 0.009 F777 VCTPVPHPI 0g.000 MYVC-TPPH 0o.099,, TABLEXII-V25-HLA-A3-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 Iamino acids, and the end position for each peptide is the start position plus eight.
STA UBEQUENCEPSCRE !F -~LPRIS I 0.0094 f 2 L RTPQj[iSS If :9 I 7 QISSID ]faooa 8 ISSIDTDPP f 0.000 1 1 FsRTPTRQIS 0.000 T ABLEXII-V25&26-HLA-A3-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified,3 Ithe length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
STARTJ SUBSEQUENCE SOE '[QISSSDT-DP [0021 I7-1' SSSDTDPPA 0.001l 4 ,1 RQISSSDTD H.01 i 47PTRQSSS 0.001 I PTRQISSSD 000 _,ISSSDTDPP [.000 7 3TRQSSSDT70.000 TABLEXII-V26-HL11A-AMMERS- I
PSCA
Each peptide is aportion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9.
amino acids, and the end position for each peptide is the start ____position plus eight. j f~TRE SBEUN C E SC6ORE I 7 4 GS-SDTDPAI 0.003 I QGSSDTD_1 0.00 '7FD 9I Tb. o 9l TABLEXII-V26-HLA-A3-9MERS-1 P S C.A Each peptidea is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9' amino acids, and the end position for each peptide is the start position plus eight. lE 7 TTRQIGSS 1[ 0.001 KILL SSDTDPPAD 10.0001 3 RQIGS o F- -GSDTDPP5 6 TABLEXII-V27-HLA-A3-9MERS- i 3PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is [specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start ____position plus eight.
WSART JSUBSEQUENCE 1
SCORE:
41SRGQALRlRA 11.00 77 R.GQALRRAQ *0000qq 1 5ABEXIII-1-LA-A3-1OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified,' the length of peptidle is 10 amino i acids, and the end position for each peptide is the start position plus nine..
[STAR I SUBSEQUIENCE SCOREI-- 7~V& LTVlSK 12.001 j18_ GTALLCYSCK 3.00i [12 1GLALQPGTAL 2.700 f106 ILALLAG F1.800 VKLALMAGL- 1.3I-50 Liii LLLWGGQL .810 5 7- G-qLLTVSKGC .675 F- 14 ALQPr.TALLC 710.60 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXIII-VI-HLA-A3-1 OMERS- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
1_ SaRT S.UBSEQUENCE S CORE I 1[CLQVENCTQL 0q.600 Q LQGTALLY 10.540.
1__21 LLCYSCKaQV If_0.200 I DLCNASGAHA .060~ 1 L69k: 1LNOVDSQDYY 11.060 J f LLMAGLALQPjj06: .iLRIAVLTVI 0.060 _gF VDDSQDYYV 1j0.060 jF Ic i LQAALAL 0~.054 LTVISKGCSL ,1 0.04 7,IFALLMAGLALQ i 0.045.
[L~L Q1 0045_ 4 ifTQLGEQCWTA 004 29 LQVSNEDCLQV 10.040 ~f 49j L WARIRAVGL ,03 ;F21 KALs VL A 0.0271 If 63 1[ NITCCDTDLC 10.020 67 SLCVDS QD 02 f72 ,IDDSQDYYVGK 101 f6 [LALLMAGLAL 0.018 F .2U.JKAQVSNEDCL 1 0.018 f _AILALLPAL 0.013 f 8 LLTVISKGCS 0.0121- FTE AL 0.009 r-7jilK TALLCYSCKA J00 f. F- AVLLALLM AG. Fj 0009o F q LNV DSQD 1000 Fj TABLEXI--HA-AM-OMERS- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peplide is the start position plus nine.
L 6- GAHALQPAAA 0.006 10. WAILALLPA,.,][ 0.q06 820 ITOD 0.005 1 I TARIRAVGLL 0.01 41 F CTLGEQCI I .0051 j107 ILALAGL 0.004 L~L~i~~Vo 0 0.004 F 5 GEQCWTARIRI 1.047' K4 EL.QCWTARIRA F .O 88 DTDLCNASGA 0.003 105 7ALLALG ~f00 F 0 FTVISKGCSLN '10.003 85 TCCDTLNA [0.003 6:2 F ISKCLC F0.00 IF 84, fT CCDbTDLC T0.002 22 FLCYSCKAQVS 0.002 40F- NCTQLGEQCW j0.0 F FLGEQCWTARI_ 0.002 [32, fNEDCLQVENC 0.002, 75 QDYYVGKKNI[ Io-.002- TK _2,ILYcTARV 0.002 F '941F ASGAHALQPA_. 0.002 IF 1 LQVENCTQLGIL0.001 ,1 10 F MAGLALQPGT ';10.001, f97 !F AHALQPAAAI !10.001 53 IRAVGLLTVI1 0.0 I _0.001 f28 AQVSNEDCLQ' 0.001 RAV GLTVIS 0.001 F731, INASGAHALQP!10.001 [7W' PALGLL\NG 0.001 1-6 IQPGTALLCYS _F-OQ-t2 F I.FARIRAVGLJT1999 1 F _7 SQDYYVGKKN F0.900'i 154 ,ITABLEXIII-V1-HLA-A3-IOMERS-_
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
START7 j UB S E QUEN CESCR ,MKAVLLALLM 1 0.0001 86 CDDLCNAS 000 ic YSCKAQVSNE 0.0001 30 VSNEDCLQVE 00 11 PALGLLLWGP 0.000 26 CKAQVSN EDO 0.000 95 S-GAHALQPAA 0.000 76 YG KKNIT 0.o,000 39 ENCTOLGEO 0.000 F11-3- LGLLLWGPGQ 0.000 [89 {TLCNASGAH 0 000 11 GLALqPqTA 0.000o 4'C bLQVENCTQ 0.000 56 VGLLTVISKG TABLEXIII-V4-HLA-A3-1 OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8' each start position is specified,' the length of peptide is 10 amino acids, and the end position for each [peptide is the start position plus nine.
S TART S UBSEQUENCE SCORE! 1153 _HLFPQEAFPA .4.500 46f TLNPVLRHLF 113.000 f7~~7iRLWAPLQPT 1,8 95- TMYVCAPVPH 1.000 11 MTHRTTTWAR 060 15-0 VLRHLFPQEA 0.60 _83 IrvLvPEAHPNA j 0.450 6 GWQASPL 0.405J 108 P"-M-ALSRTR R- 0.400 93 E SMYCAV 0,300 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 ITABLEX11Il-4-HL11A-A3-1 OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is tle start position plus nine.
[STRT SUBSEQUENCE] SCOREi F627 1[TL GVVPQASV f 30 .ITTP.YARRTSR 110,200 VVPQASVPLL 0.180 [Tq4 J HPDPPMALSR][jI 01 E[D 44q§PCSGPASYR 4o 6-090 i 0-090 [1 7 ASRQARR 100801 73 I LTHPAQWEP106 [2 TPAGPMPCSR 060 130 1f GPSNPLCCCF [f0.06 01.
fTTWARRTSRA 100501 IF -iL~ vvP~ R 0 0045 -5067, ASYRLWVGAPL 0.045 169- S-Q-VWSVVSPA 094 1 4 ACCSGDPS 0.040 167I I-DSQVWSVVS 0.F036 A TTTA [0.030 10C2 j VPHPDPPMAL 0.027 CKh7 CFHGPAS7 0.02 13 5 LCCCFHGPAF F00-20,1 39 ,SLHSACCSGD 0. 020 56 L -GA-PLQPTLGV 0,0181 1 34 PLCCCFHGPA 0.018 11Ll RTSRAVTPTC f0.015 74 1-j, LTHPAQWEPV 0.1 113 RTPTRQIGSI 0.013 I 5 fQEAFPAHPIY if0.012 If A WEPLPf 0.010- L 44 f FSTLNPVLRH 0.009 17q. SPAPSRGQAL f 0.009 1 85 if V-P-EAHPNASL 1Pb66, II6 fQPTLGVVPQA if 0.009 14 fGPAFSTLNPVl i0.009..1 TABLEXIII-V4-HL11A-A3- OMERS-
PS.CA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptide is the start position plus nine.
STRT SSEQUENCE CR L 45J TqSGDPAS YRL I0. Ta9 100 APVHPDPM 00 887 AHPNASLTMY_ 0.006 28j 7!_PCSRLPPSLR 6 27MPCSRLPPSL 0006 1 -84 LVPEAHPNAS 0.006 J zF72 1PLTHPAqWE 0O.00 5 APAGPMPCS 0,005 159' AFPAHPIYDL F0,004 1 2 f THRTTTWARR 0.41 143 f-AFSTLNPVLR 0.004 89 F HPNASLTMYV *f0.004 177 fPAPSRGQALR [0.004 1M.. LRHkLFPQEAF 036 4i 2 fPAFSTLNPVL f0.003 66 ifVPQASVPLLT if0.003 173 fSVVSPAPSRG f0.003 71 7fVPLLTHPAQWV 1f 0.003 7 if VCqAPVPHPD [0003 f121 S SI DT D P ADG;fO0 03 f 8 PLQPTLGVVP 000 F 156 4PQEAFPAHPI I 0.003 L-_5 GMC -SRLPP :f0.003 138 I CFHGPAFSTL 0.003 67 4 -PQASVPLLTH f0.003 59 ASPLWhIA 40.002 117 17RQIGSIDTDP 0.002 F. Lq ,1NASTYvcA [0,002 701.FVPHPDPPMA 0.002 32 _LPPSLRCSLH 0.2 ~i7 S .PLLT_.PAQ 21U TALEII-V4-HLA-A3-1 OMERS- PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each Ipeptide is the start position plus nine.
ThLI UBSEQUENCE SCORE: F76-: HPAQWEPVLV 0F.002- 164 IYDLSQVWSJF 0.0-02 16 YDLSQVWSV 0I0F 59 LQPTLGVVPQ 0.02 87 EAHPNASLTM 002 [92 1 ASLMYVCP .P01 [4 _NPVLRHLFPQ 0o.01.
23 4 PAGPMPCSRL- 0F0 LA7 PLQPTLGVV if0 0.01 161 VTPTCATPAG' f0 0 6oi I if- 41 fSACCSGDPA 70.001 115 I7FTQIGSIDT 0___00_ 4 12 [4TS-RAVTPTCA, 01 4 WARRTSRAVT 70.001 ITABLEX1II-V19-H11A-A3
IOMERS
PSCA1 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide isI 10 amino acids, and the end position for each peptide is the start position plus nine.
FTRSUBSEQUENCEISCORE1 8 RLLPSLRCSL< 1.350 FLI-PSLRCSLH 0.200 3 11 PMPCSRLLPS 0.012 4MPCSRLLPSL ;I0.009J 5 CSRLLPSLR F_0.0-0 10 PSLRCSLHS 0001 AGPMPCSRLL i1 0.000 7 SRLLPSL-RCS 0.0001 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXI II-V20-HLA-A3-I0MERS-' I PSCA Each peptide is a portion of SEQ ID NO: 8: each start position is specified, the length of peptide isI amino acids, and the end I position for each peptide is the L start position plus nine.
[STA RTr SUBSEQUENCE'I SCORE! 2 I~CCSGDPASSRj_0.0901 7--7 ASRLWGAPL .j09; [771771CSGDPASSRL 10.003 6 PASRLWGA 0.003 LI A CCSGDPASSJ 0.000 [q7 GPASSRLWG 0.000 L7 SSRLWGAPLQ 0.00 4 GDASS RLWl 06.00 B LEX-V21 &22-HLA-A3- 1MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 1 10 amino acids, and the end position for each peptide is the start position plus nine.
STARi SUOBSEQUENCE! SCORE: JE 7LtDLAQWEPV v.010 3T FSVPL LTDLAQ 0,004O- IF1 QASVFPLLTL f002: 7Y-[ DLAQWEPVLV f-T001 F 8if DLAWEVIL [0.000 IASVPLLTDLA 10.000 [PLTDLAWE ~I 0.009 fTABLEXIII-V22-HLA-A3-1 OMERS- I PSCA Each peptide is a portion of SEQ ID, NO: 8; each start position is specified, the length of peptidle is 10: amino acids, and the end position for each peptidle is the start position3 plus nine.
[START SUBSEUENCESCORE I 9HL QWEVLV I 0.200 LLHLAQWE V 0.060 =~PLTHL 0.009 [77f7VPLLTLAQW 0.005 ~FT].SLTH AQ IF0.004 5PLLTHLAQWE 0.003 8 THLAQ(WEPVL 0.003 2 ASPLHA 0.002 1T 0LQ E VIP 0.002 TABLEXIII-V24-HLA-A3-1 OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position Ifor each peptidle is the start position plus nine.
T SUBSEQUENCE3SCORE 5 TMYVCTPVPH 1.000 3 TSLTMYVGTPV I 0.300 1 [TPVPHPDPPM [0.007- 7 FYVCTPVPHPD[0.003, 4 [LTMYVCIPVP 0.002 If2~[ASLTMYVCTP. __ICTPVPHPDPP 0f .001 F 1-1 NASLTMYVCT jf0.001 F8 1VCTPVPHPDP 0'.000' jf6 MY-VCTPVPH-P 000 TABILEIRll-V26-H LA-3IOMVERS-
PSCA_
Each peptide is a portion of SEQ ID NO: 8; each start position is Ispecified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
3-7 RTPTIRQISSI 0.4 [77[-7q~5PD 7 .0 9 I T S-DTDPA .00 ITABLEXIII-V25-HLA-A3-1 OES PSCA Each peptidle is a porion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the and position for each peptidle is the start position plus nine. I START S UBSEQUEN CE, CRfj 5 jPTRQISSIDT_ Jff oo jL 4 I TPRQISiDJF0.000 J 1i S:RTPTR1QISif 10.000 2 S[RTPTRQISSI0.0 6_ TRQlSsIDTD If670003 TALXIII-V25&26-HLA-A3-
IOMERS-PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 101 1amino acids, and the end position for each peptide is the start position: STR plus nine.
6 .1QISSSDTDPP 'f 0002 7 [_SSSDTDPPA4 0.001 IRQISSSDTDP 0[ o ooT K I TRQISSDT 0.001 P IJTRQISSSD I o.ooo 7Y SSSDTDPPAD .0 1 .4 TRQSSSDD if0.000 ,TABLEXIII-V26-HLA-A3-1 OMERS-
PSCA
Each peptide is a portion of SEQ ID NO.-8- each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
fSART UBSqEQEE SOR s QGSSDTDPP 0.002 3 PRQIGSDT~f0.001 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 GSSDTDPPAD 10.000- [77J6 qfSSDTD 0.000 4 TRQIGSSDT 10.000 TABLEXIII-V27-HLA-A3-1 OMERS-
_PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position ,for each peptide is the start position ____plus nine,.
[STAR SUBSEQU ENCE SCORE PSRGQALRRA f 0.000 L 2 RGQARRA .0.000 [TABLEXIV-V-HLA-A1 101-9MERS- K PSCA TEach peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position ll -plus eight.-,- F SU-BSEQUENC SOR START E [CR f'K7 SQDYYVGKK [0600 19 TALLCYSCK If0.300 -F GTVISKS 11[0.060 FOLOVFSQDYY IF 0.020 r'y KAVULL.AlLM 0.018 A 'qfGLLQPGTA [0.012 Y~ALLi 0.012 f2 fAQySNEDCL 0~.009 if 54 AGLLTl:VI uog ]66 If. 47 QCWTARIRA 0.008, F43 QLGqOT .0 [TABLEXIV V1-HLA-A1101-9MERS- Each peptide Is a portion of SEQ ID, NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight If.SUBSEQUENC 1
COE
109 ifLLPALGLLL If0.008 L115if jLLLWGPGQL .006_ 17 1 f t LALALG =.006 20 '5 FALCCA 0.006 I f GTLLYS 0 lREEN 006 1 'if CAI LALL 0.006 83 N l TCDTDL 4:~0 Q:11 f LAGLW 04 I I L L FdjIEif miF4 -E QWT IfbPAAAILAL 0004, 14 ,r fLQTALLAL F 0.0043 69 EQCWDSQM 0. 0043 8 FHALQPAAAI [0.003 52 IRAVGLT 0.002 29 QVSN EDCLQ O TARIRAVGL 0.002 I VG LL ViS_ 0.002 37qf 7 VNTLG J 0.002 86 Ij C C DDLCNA 0.002 I GLLTVISKG 0.002 114 GLLLWGPGQ 0.00 TABLEXIV-V1-HLA-A1 101-9MERS-!1 P...SC A Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position Ifor each peptide is the start position plus eight.
START SBE ENSCORE I454GEQCWTAI Ef 002 I L VLALMG f00 D 0 ~LCASGAH [10001 LALGLLP 00 F -8 4L A L L P oi f 00 F'if7Y7LW7GKNT] .F-0c T-65 [GCSL-NCVDD) 000.q1 -I 7K v-s NE6b' 0~o1 30o FvSNEDCLQV 0000~o F1Y_-:7CLQ-VENCTQ 0000T 4F 7 VDDSQDyYVj 0.000 S1 7IKGSN7 0000 1 0 I7T3LLP AG 0000 8-5 4TCCDTDLCN f0.000, T, 92 ONASGAAif00 F 53 F IRAVCLL-Tv [00006 7[I 6-YsciKvs- 10.000 1F 67 1SN DDSQ f0.000~ T- r8 DCNSA 0.0001 1 TD 0.SG[000 51,- FARIRAVGLLif 0,000 j 8 LTVniSKGC Fi000066 40 4 NqTqLEC i.00o 3L s SKCLNCv q00 :F 93 NASGAHAL-1Q-if0000 F4- 10,[MGLALQP G [766K WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXIV-V1-HLA-A1 101-9MERSj C. Each peptidle is a portion of SEQ ID NO: 2; each start position is speified, the length of peptide is9 amino acids, and the end position for each peptide is the start position plus eight.
START !SBEQUNIS-CORE, 2 LCYSC0KAQ~ 0.0 00 I CAQSE 0.0 ES:GAHA-LQPA I[0.000 ,LLJA~1vLL LLJ[k9 [I:AAAILL 40.000 L82 KITCCDTD [0.00 it__38] VENCTQLGq _0.000I 347 DCLQVENC 17.ooo 64 -jKGC-SLNCVD 0.[0 111FrALGLDWG 0-.0007 3j2 NE DCLqVEN, FO66 6BLNCDDSQD- 0p.000T 94 L[ASGAH.ALQ.P o.o [TABLEXI V-V4-H LA-Al 101-9MERS-'
PSCA
[Each peptide is a portion of SEQ ID; NO: 8; each start position is Ispecified, the length of peptide is 9 amino acids, and the end position for each peptide is the Start position ___plus eight.
SATiSUBSEQY ENCSCR 13ISVVSPAPSR .0 iff k6 [TWRR TS F[0.400- 'F'19 I MALSRTPTRf 0.060 f 1781 PSRGQ 6L 1 0040j f145jSTNPLR 0.0301 84 I LVPAHPN if0.020 L ILTMYVCAPV 0.020J TABLEXIV-V4-HLA-AI 101-9MERS-1
C..A
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 91 amino acids, and the end position for each peptide is the start position E -Plus eight.
S UBTSSEQUENC 70j VFLLTHPA [0.020 1L TTP j[0.-0 10 K 1 ILTRTTTwA 10.010 I 64[GWEqVPQASVF 000 11 [RTPTRQIGS 0.006 L JAP QPTLGV iL00 96 MYVAVHfT06 4 7V F RLWGqAPLQP 0.005 7,Vi[SVPLLTH j__ 160 PHPYDL[0.004 51 'T7-yL~wGAPL La.00 4 29 P CSRLPPSLR j 0.004 F7Y ''PAG-PMPCSR 40.004 1CSGDPASYRl 5 0.004 14f STLNPVLR 0.004i F- ii iL TSRAVTPT 0.003 F 163f HPIY-DL§qV ay 11 RISIDTD 003 F78 FA [l\L7 0.002, 1146ATPIGS '70.0021 15 AV PTPP9 06.002 T7EL YVC VPHP F0.002 F 6 FYP( I -VPL~ 0.002 42 SCCSPA S0.002 1j 43 AFS--TLNPVL'00 102 IvPDPPMA [0.002 [89 HP NASLTMY [qf:002 TABLEXIV-V4-HL11A-A1101-9MVERS- Tij Each peptide is a portion of SEQ ID NO 08; each start position is specified, the length of peptide is 9 Iamino acids, and the end position Ifor each peptide is the start position plseight. START S QE CO RE 17 SPRG QA 0 002 101 VP--D----0.00 [44 CSGDPASY 0:.00 F 174 7 FWS-PAP SR G 0.0 61 K-PTLGvvpQA 0.0 141 GPAFS-TLNP I01 158 7EAPHI 0.001 17 DLSQVWSVV 001 J1206i 7G IDTDPF 0.001 T152 4 RHLFPQEAF f o0-0oi 1133_.~FNPLCCCFHG if0,0 f169 if SQV-WSvv-SP. 01 F 14 3- IqT TCA-T7- 0.001oT ;F149 F- VLRHLFPQFT0,001 81~ T IEVV66.H 001 1179 PRGQAL5RRJO.o01 J164-1 -IDLQ[ Ifay 0,00 0.00 1 153 HFPEF 000 t56i' rGAPLQTL 0.001 L' I LQPTLGVVPVdi1 [966: DSqVWSF-6 00011 37 RCSILHSACC 10.001 148ji~ NPLRHLFP f 0,00 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 ?TALEXV-V-HLA-A1 101-9MERS-'i
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
STR SUBSEQUENCi CR 138TJI CFGA S 0.00 muO. IiNEVLVPEPA_ 0.001 917 NASLTMYVC =1 0.000 Li2JL] S I DT P c07~ D ib NOR yLHLF -PQ E F 0.000 147]LNPVLRHLF -0.0 0-0 L_13 ]fCCFHPAF~if0.000 118 iQIGSIDTDP o.oooi 46L3- fDa YRL [0,000 [w fSN PCCC F o o 53o: ALSRPTRQV fo.ooo 58 jLTLGVVA 0.000 146 ITLNPVLRHL 0o.000 73 fLLTHPAQWE KO Q, F-i. :AHPNASLTM [0.000~ [-165 'D -LSQVWS [oPoo 74-9 PASYRLWGA_[0.000 142i ASTNV .0 NO: EXIV-Vl 9-HLA-A1 101 Each peptide is a portion of SEQ ID N:8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight. J scoUN I~ KA SRL S ORE00_ 7 -7 PSLRCS 0.0 TABLEXIV-V 9-HLA-AI 101 -,..,9MERS-PSCA Each peptidle is a portion of SEQ ID~ NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
START SUBSEQUENCI SCOREI
E
EPLRSL 0.002 GPPSRLL I 0.00! 3 _.MPCSRLLPS ,0.000 4 PCRLLPL i .C00 i[ E[SRLPSCj .0 Each peptide is a portion of SEQ ID NO: 8; each start position is1 specified, the length of peptide is 9! amino acids, and the end position for each peptidis htr position plus eight.
[ISTART'SUBSEQUENCE, SCffE CSGDPASSR_ 0.004 6[ PASSRLWGA L 9 8 i -SSRL.WGAPL 0000 1- -CC~SGDPS 0.000 [3 SGDPASSRL: 0.000 DPASSRLWG 0.000 dPASSRLW i0.000 9 SR-LWVGPLQ[ 0.T000 j 7 ASSRLWGAP j 0.000 [TABLEXV-V21-HL.A-A1 101- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9[ iamino acids, and the end position for each peptide is the start ____position plus eight.
[ST;SUB SEQU ENC SCOREI 3: VPLTDP .020 ~FT7 LTPAQWEP !10.002 TABLEXIV-V21-HLA-AI 101- 91VERS-PSCA__ Each peptidle is a portion of SEQ ID NO: 8; each start position is Ispecified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight. 7 LPLLTDPAQW I 0.001 F PEAQWEPVL 0.001 L: _LTDPAQW E IF QASVPLLTD 1 056000 E EVPLLTDP-AQ. 0,000~ 8j [PAQWEPV J_0.00 :LZI ASPLDP ooo 0 ITABLEXIV-21&22-HL11A-A1 101- ~Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 91 amino acids, and the end position for each peptide is the start position plus eight. [TR{SUESEQUENCE
SCORE'
f VPLLTDLA 0,020 6 -LTDLAQWE 0.002 8 IDLA WEP_[i.o J LLDLAQ 0.001 PfLLTDLAQw 6-0.00 If7 fTDLAQWEPV_[ 000 TABLEXIV-V22-HLA-AI 101- I 9MERS-PSCA Each peptide is a portion of SEQ IDI NO: 8; each star position is specified, the length of peptidle is 9.
amino acids, and the end positionI for each peptide is the start position plus eight. [ST.Rj SUBSEQUENCE
SCOREI
F 2] VLTL 0.020 -8 1 .I-HAQWEPVL 004 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXIV-V22-HLA-AI 101- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position p__lus egt STARSUBSEQUENCE7SCORE [9 LAQWEPVLV 0.00P2 LL. 1[ :VPLLTHLAQ :[0.001T [4 PLLTHLAQW 0.01 ASVLLTHL i .0 F TABLEXIV-V24-HLA-AII101- 1 Each peptide is a portion of SEQ ID.
NO: 8; each start position is 1specified, the length of peptide is 9 amino acids, and the end position for each pepflde is the start position: plus eight.
S TART ~~iN]SCORE' [f 37 L-TMYVCTPVI110.020 1 6 FMVTPF 0.006 8 TPV PPDP1 0.001~ 4TMF1YVCTPYP' 0.001 lK SLTMYVCTP 000 K7VCTPVPHPD 0,000 I177 F ALTMYvcT 0.590001 101- 9MERS-PSCA FEach peptide is a portion of SEQ I6 NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position ~plus eight.
STA 1SUBSEQUENCEf
SCORE
2 RTPTR Q SS 000 TABLEXI V-25-H L1A-Al 1101- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start position 1ii 8 SSIDTDPP 000I TABLEXlV-2526-1-11A-A1 101- 9MR-PC Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position Ifor each peptide is the start position plus eight.
STAR SUSEQENCEz SCORE 4 RQISSSDTD 0.0 f5'. QISqSSDTDP 0.0 7 SSTDPPA 0,000 I TPTRQISSS 0j .000 TQSSSD_~ 0.000 6 ISSSDTDPP 0000 TABLEXIV-V2-HLA-AlI1 9MERS-PSCA Each peptide is a portion of SEQ I1D! NO: 8; each start position is specified the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
START SCORE!
E
;F .4[RQIGSSDTD! 0.003 7 FGSSDDP 0.001 TABLEXI V-V26-H LA-Al 101- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is I specified, the length of peptidae is 9 amino acids, and the end position for each peptide is the start position 1 I TAT UBSEQUEN L-6I[§ QIG~S-SDTDF j[ 00 1fPTRQGSS 0.000 K i IGSSTDP LO 0 8 SDTDPPAD I00 TABLEXIV-V27-H LA-Al 101 1AE~S-PSCA Each peptide is a portion of SEQ I0D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positionI for each peptide is the start position' plus eight
SUBSEQUENC'
START 'SCORE 4 E I 2 RQ LA 0.000 [TABLEXV-Vl-HLA-A1 101- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptidle is amino acids, and the end position for each peptide is the start position plus nine.
FFSUBSEQUENCi START E SCOREI 55- 1 AVGLTV-K fc 400E jf _____GT-ALLCYSCK[30 Q3 LGEQCWTA 0 R 0.080 29 11[QVSNEDCL-QVf[ 0.040 70 FCVDDSQDYYV 004 562 RIRAVGLLTV 0o.0247j, WO 2005/014780 WO 205/04780PCT/US2004!017231 TABLEXV-V1 -HLA-AI 101-1 Each peptide is a portion of SEQ I D NO: 2; each start position is specified, the length of peptide is 10.
amino acids, and the end position for each peptide is the start position plus nine.
ISUEUENCI
I[STAT U SCORE 114 I[GLLLWG PGQI.[ 0.0181 42 TQLGEQCWT 0[.018 LT-TTV ISKGCSL [0.015 12 ,EGLAQP T0.1 Lo0Fj QPAAAwLAL f01 1--PM 108 IfALAGL f0.012 106 ILAPAG! PD 0 0.08_ JF991 ALQAAALA .008 109 LLPALGLLL 008 I .LdAVLLALLMAG F *06 i 6 f LLLMAGLAL 0.006 Fk II LLMAGL 006 96 'GAHALQPA 1 0006 7[2_ DDSQDYYVKIFb.~qI 103 1AAAIILLP 0.047 F 35 [CLVENCQLG 5 .04 f~V7 5 f
LALMGL
rTALMGP:10.004.
GECTRIR [0.,0047 19 TALCSCA ii70.003i [7-60_ [7LLLPALG Ll 0.003 6 [NVDSD 0.003 (:so iLVSGSNI003 5AIALPALI 0_.003j 13 LLQPGTALL 0.003 TABLEXV-V1-HLA-A 1 01- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position Ifor each peptide is the start position I START.[ BEQUN SCORE." [84 LTOG [-DDLCN 100
L~
8 5 ~TCCTDLCA 1002 F0 C -N1dL]0.0 L if L LMA LALP .0 [77 YVGKNIT~c If 0.001 5 41 CTQLGEQ7C fooi 28 A,,[QVSNEDGL-Qj 0.001 F36 L EN QLG 0,001J GLLTVISKGC 001 A tqPTALLCqfoooi: 6 1 5VIYSk GSNC 0.001 7FGG SLP CVDDSji0.0011II iF-7 JALflMAGLALQ 0.001 F837 [N iTCCDT~i6 0.000 47 jQ§CWTARRAV'4 0.000 1 Mi~KAVLLALLM_[ 0.000 F1-10 o LPALGLLWG IF 6 66T F 9 7ULALQPG 000oo r2ILYSkA~qvs 0.000 5' 3 LbYSOKAQV SNT'.C6071, if67 IISNCVDSD 0.0 Fth24A'PGLLW P ]I110.
LLTVISKGCS 0.000 ,7j I NAGAHALQP 0.000 UTABLEXV-Vl-HLA-Al 101- I OMERS-PSCA Each peptidle is a portion of SEQ ID 5NO: 2; each start position is specified, the length of peptide is 5amino acids, and the end position for each peptide is the start position plus nine.
[F SUBSEQUENCI FS ST ART E SCORE =20[n ALCYSCKAQ =0.000 tiliAGLAL QPGTA [OU00 1 7 DYYGKKNIT 0.0 6 QPGTALLY 0.000 FT 101MAGLALQPGPCT 0.000'j II 8-67 CCDTDALCNA 0[.0001 15 75C QDYYVGKKN 0.000 1 957 ASGHALQPAlo 000q F25 ['SCKAQVSNEf o.000 34 jDCLQVENCTQj 0,000 51 [AV L:TiF .000 -1 ALLLG 000 f-64 lFGCSVDD F0.000 !5 4Z NE6dLQvENO';'- 0.000 92 FcNASGAHA1T6 o[co 31 SEDOLVEN 0.000 1.TABLEXV4-1=HLA-A1 101- WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Each peptide is a portion of SEQ ID; Ii NO: 8; each start position is specified, the length of peptide is 10, Samino acids, and the end position Sfor each peptide is the start position; plus nine. SjTART SUBSEQUENG SCO RE F 1 MTHRTTTWAR
DQ
f64 GPQASVPL E000 I 178 AP SRgQALRR 0.080_ F 4 1CSDAY 0.040 1 TPTRIGSI I fHLFPQEAFPA F0.0241 F 15 FATGAL _T P F 0.020 6 ITTWA RRTR 0.020 [7 VPSVLI 0.020 _T31 RLPPLJ .1 sIfGA PLQPTLGV{.W912_ K74 LTPQEPV 010 169 SVSVP o.6ooPl jf 108,F P-MALSRTP TR I b7.-9bY- 837 1 LPEAHPNA!R 0.006 1F 30 IGPSNPLCCCFiI 0.006 IF 41fGA-FSTLNPV 0.006 62 fTLGVVPQA§V.I 0 .004.
S146F TLNPVLRHLF 0.00, q4 i170 dQ0VWSVVSPAP 0,004 1102, VPHPDPPM1AL-- c.104, 1,7 APRG AR 0.004 17F_ VLRHLFPQE 1!000 PCRLPSLR .10.004 IF7 sL-m~yCAPv!4 0.004 159 AFPHPIYL 10.004 VA I~O00~ JTABLEXV-V4-HLA-AI 101-
IOMERS-PSCA
~Each peptide is a portion of SEQ ID: NO: 8; each start position is spec ified, the length of peptide is 10~ amino acids, and the end position Ifor each peptide is the start position lus nine._ rSATSUBSEQUENO SCORE, F1_0_0 APVPHPDPPM, 000 JTPIQPTLGV[003 IF JLFBSR rT91 0.0031 L THPAQK1 0003 1L7 R~QIGSI DTD_ 005:63 5 E _MPSRLPP 000 F Aq RLWGAPLQPT 000 43 ACCSGPASY~0.002~ jF91 ,[!NASLTMVA[F 0.00 T 1 CLAPV 40.002 84_ LVPEAHPNAS .0 HP; QWEPVLV f o.ooT, 32~ PP SLRCLHLY6 07 60 9~PTLGVVPQA i 0.002 .1 5 jVEAHPNAS[02 F__10KFPVPHPDPP~ 0 15 fLFPQEAFPAH 0 0602, ii_15 ~IPAPRGQAL' 000 1138 ICFHGPAFSTL 0F_6.002 1457 STLNPVyLRHL l00E2, 1137 4CCFHGPAFT ob7 F 21 FI[KiPGPmWCSD 0,001 _148 IrTGATPAGm! 0.001 1Lj4., [N5PVLRHLF0X01 11 TABLEXV-V4-HLA-Al111- I 1OMERS-PSCA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 10'1 amino acids, and the end position for each peptide is the start positiont plus -nine.-
~SUBSQUENCSCORE!
SET1RTCoE 0,001 37 RCSLHSACS 0001q~ 1 180 SRGqQALRRA 000 NiSIDTDPPADG f0,000 -f.14 7 7PLCC-CFHGPAf 000i 16 11VPQASVPLLT if0.000 K[144 {FTLPLRI00 'F--718 1 1 Ti5TP P I ~x IF 142 PA_,FSTLNVL.I 0.000 S51 SLRCSLHSAC'[0000 5._71 _1SYRLWGAPLQI: 0.000 50 ASYRLWGAPLI 0.000 39 SLACSGD! =0000 110 ALSRTPTRq .0 145 [A.DPASRL 0 69 ASPLLTIIRA i0.00 F' RT TWA RRT S~ 0.000 13 DTDP PAD GP S 1 O_.0_0 [7F 16 FYDLSQVWSVV. 0.000 F 2 82 PVLVPEAHPN 0.000 f4j13 NPLCCCHG 000 F7{4§ PVL(LFQ 0.000 -6 L_LSRPTRFqI -0.00 TABLEXV-VI 9-H
IMERS-PSCA
'Each peptide is a portion of SEQ ID -NO 08; each start position is WO 2005/014780 WO 205/04780PCT/1§S2004/017231 specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position _plus nine.
[TR SUBSEQUENO E SCORE! [7 81[RLLPSLRCSL 0.018 jIPCSRLPSL If0.0047 iF i9.. j LP--Es-LRcsLHS I[ OTQD Im~pC-RLL s F I 0 1 G RLL I[ 0.000-6 I7 1S-RLPSLRCs:[ 0.000 LA-All 01- I[ IMERS-PSCA- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amine acids, and the end position for each peptide is the str pstion plus nine.
START!'ISUBSEQUENCE4 CE. 50 2 -0GPA I. 0 40' 8 ACSRLGPAPLS 0.000 3 1 CSDPASSRL 0.000 GDP TASSRLWG 0,000 F, 760 FSRLG9A-PLQPI 0.000 4 [I-sGDPA SLi0000 -71 FPASSRLWGFI F9 S-SRLWIGAPLQ 0.00067 ,_TABLEXV-21HLAA1I Each peptide is a portion of SEQ I D NO: 8; each start position is ispecified, the length of peptide is 10! !amino acids, and the end position 1 for each peptide is the start Position; plus nine.
jSATSUBSEIQUENG SCORE L 8j7 LTDP!AQWEPV 0.010 [~7VPLLTD PAQW I 0.003-- EtIV SVPLTPfAQ 0.002 II7LLTDPAQWE J O.0O1 1 DPAQWVL V 0.001 I__jASVPLLTDA00 9 17TDPAQWEPVL 0"000 2 QASVLLTDP 0.000 TALX-V21&22-HLA-A11Oi- 1 OMERS-PSCA SEach peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
ISATSBSEQUENCIESc ill F FSVLTLA P0A 1 4 VPLLTDILA IW jf_0 003 QAsVPLTDL [0.002; 7_IF 5JLcQE L [0.0011 II6 LLDLAWP 0.001 II8 DL EPVL][OO V2 ASVPLLTDLA, 0.9909 TABLEXV-V22-HLA-AI 101-
IOMERS-PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
I TART SUESEQUENSI IF I E SOR 1 7 LTLQEPVj 0.010 L 9 HLAQWEPVLVF 0047 [I VPLLTH LAWF 70 04 t TABLEX V-V22-H LA-Al 101- Each peptide is a portion of SEQ ID' NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
[START SUBSEQUEN, SCORE I, =[§APLLTHL[q 002 F c TLQ(WEPVLP[f 00 2 4-ASVPLLTHLA IF- o=oooi ,F [PLLTtLA QWE]I000 TABLEXV-24-HL11A-A1IO1-
IOMERS-PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is Ispecified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
[fXfjfSUBSEQUEN CEII'SOOREI I-7 TMYVCITPVPH,.Il 0.0081 F[77 SLTMYVCTPVI E1OT\/HPPM 0.0031 P4 TMYCTPP 0.002 F77YVCTPVPHPD Ii 0.002J [PcThvPHPDPP. 01 6 7N YCT PPP i 01 Q 2 7ALTMCTP[Y000I1 IfTABLEXV-V25-HLA-AI 101- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position~ plsnine.
START,[SUBSEQECE ISCORE 377 RTPTRQISSI 0305 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 101 D MERS-PSOA___ L T7RQI1SSID TDP I o-003: 9 ISIDTDPA 00 PsTRQISSI T ooJ Ff STPTRQSS 0000 F7IJJRQSSITD 0.000 1 fLSRTPTRQIS 0[.-000 TABLEXV-V26&26-HLA-AI 101- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end Position for each peptide is the start position plus nine.
F:STARTSUBSEQUENCE1 SCORE LI I'-,[RTPTRQIS -1 If:1 Dq 1F- 5 QISSDTIP I[o.o03 6 f QISSSDTDPP. 0.000 71 ISSSDTDA_ 0.90 F2- TPTRfG*SSSD7 0.000 1F37 [PTR:q-lSDT 110,000j TABLEXV-V26-H LA-Al 101 1IOMERS-PSCA Each peptide is a portiono E ID NO: 8; each start position is specified, the length of peptide is 1D amino acids, and the end position for each peptide is the start position plus nine.
4 SUSEQUNCESCORE; 1 1 RTPTRQISS 9 .003 1_5ILRQSSDTDPJ0.3 K_7FI SSDTD PPO 00-0o 2 ,FTPTRQISS-SD -0,000 3 PRQIS SSDT 0.000 4 TRQISSSDt 0.000 EC SSD TPPAD 000 TABLEXV-V27-HLA-All0l- 1 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plsnine START I SUSQb l -CORE 2 iSR-GQARRAJl 0.00 TBE V1V-HLA-A24-9MES Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start ~position plus eight.
1JSTARISUBSEQUENOE
ISCORE
1 4 76 DYYVKKNI 50.000 I 7 [YYVGKI 9.000 36 LQENCTL ,f7.200 T 99 ALQPTALC7.20 109 A LLPALL 7.200 60 FTVISKGC~SL 6.00 F-2S3AQS 6.000 F 7 ALLMAGLAL [6,0 I- T5 FLG'PGQL j "6.000 F2-~ 7AQVSNEDCL [Tooi -17 LALLPALGL 6.000 5 -L LLMAG1 m l 4.800- 7 11FQPAAA-ILAL iF4.0700 8-63 1 NITCCDTDL 400001 57 TARIRAVGL 00-0 9 FY:NAGAHAL B .000 TALXVI-VI -HLA-A24-9MERS- 4 PSCA Each peptide is a portion of SEQ ID NO: 2; each start position'isQ specified, the length of peptide is 9 amino acids, and the end position for each peptide is the str po__Rsition plus eight. F-TRT7 SUBEQUENCE SOOREI [54 RAYGLLTVI 7 if 98 [ELQP-AAAI 1.500 7 ii4 MKAVLLALL i ,8 117--T QVSNEDC 0.30 1 34 fDCLQVENCT 0f.25 69 fNCVDDSQDY :f2t6 30 .[VSNEDLQ F 0.180 F20 [0LYCA~f.166i o FGEQLicWTAI jis TQ LEQW if0.150 3 AVLLALMA- 10150 FT100 4 LQPAAAILA rO.150 i r66:rCSLNCVDDS 0O.150 10-4 M-ILALLPA 0.1 5 0 1.-9i ]FCNASGAHA 'F67J57 Fg58' 1LL"T'V SKGC f'140 55 ~7 AVGLTVIS__0.120 1157 TCCDTDLCNFoiW _431 QLGEQCWTA 0.120 1'_GTALLOYSC 402 N C T Q L G E Q 4 0 .1 2 0 1 96 7 fGAHALQPAA 012 L~ VDDSQDYY 110 F151 SGAHAQAI02 ;F 61 VISKGCSLN 010I WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXVI-V1-HLA-A24-9MERS-
PSCA
Each peptide is a portion of SEQ I C NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start ___position plus eight. fTTSUBSEQUENCE' SCORE: 'F471[__QCWTARIR [110.1LO 1 II[ QA LLGK -I -0.100' -84 jTCC; DTDLCq F -6jo 186- CC-(DTD LCNA 000 [481 CWARI RAV 0.760 1 791 yGKKN- IT(;c- 0.100 4 82 L~NICCDT., ~0.030 KGCS-LNC( VD [0.024 GLLT-VSKGO02 31 1 S..NEDCLQVE il0.022 [67 S0.021S F11 57 F GLLWGP Gq-F 10,01 F37 1[QE TLG:0.1 I YLLLL A 10.018 73 DQDYVGK 0.018 1 [-87 1[ CDTLCNAS 0 17 I QDYVGKKN Q 0 7T LFEQCWTAR 0.015 'F19 T ALL CCK 0. 0 1 5 If 114 [GLLLWGPGQ 0.06Ts5 S][TDLCNASGA 0.015 I VGLLTVISK 0.015 L 3511 [LQVENCTQ, 0015 497 WTAR IR-A VG- 0.014 f3 327 FNEDC L-QVEN- 0.013 '103 F ALLP 001 112 q ALGLLWG 0.012 TABLEXVl-V1-HLA-A24-9MERS- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start ___position plus eight.
[START S§UBSEQUENCE SC OR f9 [LMAGLALQP [o,12] 2MGAQG iTABLEXVI-V4-HLA-A24-9MERS-
-PSCA
Each peptide is aportion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9: amino acids, and the end position for each peptidle is the start position plus eight.
START! S UBSEQUENCEI SCORE] M, YV CAPVPH E: 0.750 1771 PASRGQAL 0.72 17 PGTALLCYS Ii 0.012 §94 JQ YYGKK 5[0011] 7 QVSNEDCLQ ]fo 25 SKQVSNE_ 0F.010 TABLEXVI-V4-HLA-A24-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9j amino acids, and the end position for each peptide is the start __position plus eight.
:1511~Y~iIG 200.0004 L1~,.LZ~TLPVL [24.0 317 PCSLRCSL T7.200 55 VPQSP 6.000 I, 66 7VPqASV L, I600 724.47 AGMPCIS RL 6.00 55 WGAqPLQPTL 5.7f60 16 57 IyDLSQVWS 5.000, 46 SG-DPASYRL-1 4.800 P 160j1. FPAHPIYDLJ4,000-- FJ 6 F 7HPAQ-WEP\VL .14,000 I [136i LN PAHFI[2.000 114 -fTRQGSI_ j1.000 :1 11 1LRTPTQI 71.000 154FU LFPQEAF 07506 1, 12 LRHLFEA 10.600 6i F FHGPAFSTLi F40.76 I P-CSR LPSL .48ao EL 2 7 FP AD G PSN PL7 11 O.480 131 PNLCF 032 1 R L P-PSLRCS f. 67 I_147 R-AVTPTCAT 0.300q7 94: LMYCAPV I 0210-L J9{. RTSRAVTPT 0.200j 4 _RTTTWARRT 0.01 37L RCLHSACC 84_T LVPEAHPNA__ 0.1780 [8 F W 7 E-AHPN Ifo0.180§ 785 1 PAPA 0.180 f 7 Y S P L L T H A II 0. 8 120 ,GSIDJDPPA~O~ S62 7[TL6WPQS~ 0.168 7~7 PLQTLGV bF.io[ 7-16 17i2 j WSW--SPAPS 10,150 T-1635 HPIYDLSQ 0.50 129 DGSPLC40.150 F 7.HGPAFSTLN 0I .150 16 i4PPM-ALSRT F 0 .1 i 89 HPNASLTMYJi[o1iso F 7 4 DP PiDGPSN~ 4 1 is CSLHS ACCS 4 0.150 _1 V-TPCATPAi[0150] WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXVI-V4-HLA-A24-9MERS-I PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptidle is the start ____position plus eight.
[START SUBSEQUENCE [S(CRE I F92 A L TMY VCA .5 QVSSA 0.140 'L!677 DL SQVWSVV 0.140 12 TSRAVT-PTCJF 8 EAHPNASL 0.12 I158 Ir EAFPAHPIff 0A1 L76 JFATPAP§RGQ2L Lf.29 F02:]F P HPDPPMA jfk 0.2 27 M[-PRLPPS_ I0.100 7 T wARRTS-RA__ 0.100l 2j[ TPAPMPCS ,0.100 [81 [WARSRAV 0. 100 F .1ACSGPPAs :10100 E IFTTT RT 1 00 9 THRTTTWA 0.100 IFW: CSGDASYF.100 1357LCCHP 0.10 LisT, -QEAFPAHi [0.100 0.075 [767IF PPHPDPM 0075 1103 PHPIDMAL_'L6'697 86 FPEAHPNASL 0.040 F181 -iRGQAL-RRAR 0036 1717 RQIGSIDTD F q 0.03 D1E~s,. jAVLTH 0X1L0.Q22 liIsF71 FPQEAFPAH 1022I jTABLEXVI-V4-HLA-A24-9MERS-
PSCA
Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position i for each paptide is the start position plus eight.
'FT)EfSUBSEQUENCE FSREJ D67 ILPTLGVVPQA j[0.021 E1 PVLYPEAH H21~~i 5[ RL GAQ 002 80 W[EPVLVP PEA.:[0.0201-1 124 TfDPPADGPS I0.08 58 PL Q PTLG-w 0o.018 30TO_ SR-FL PP SL RC 0.018 iDq64 f GvASV if .oii-i F16-4,-4 PIYDLSQVW'f.017j ~f98TVCAPHD o.1 TBEV-VI 9-HLA-A24-9MERS-! __ABEXI PSCA__ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 91 amino acids, and the end position for each peptidle is the start psition plus eight.
IKT RT SUB EQUENCqE SCOR
E
G. FP.MP..SRL. 7.1200 8 LLSLRSL 7.200 K[7 -RLLPSLRCS 0.360 3 MPCSRLLPS ,100 6 SRLLPSLRC -0 0 1s 5 CSRLLPSLR 0.012'' F2 F PmFPSR LP F 0002 -ME'S1 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 fo achn puthe is the start JsIART-I SUBSEQUENCE. sCOREJ 37 SqDPASSRL EL±001I if 1 [CCS ,PASS foio f 77] ASSRLWGAP 0f.0~ 6 PSSRLWVGA 0.010] 5 DP ASSLG 0.010 JTBEV-V21-HLA-A24-9MERS- IT1LXIPSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9! amino acids, and the end position for each peptide is the start position pius eight. START SUYBSEQUE.NCEjCOREJ, 2F C 1 S VPLLTD 0,022 5 PLLTDPAQ 1 0.015 [LLTDPAQWE10,4 7 ,[LTDPAQWP16U 1 Q SVPLLTD 0.010' TABLEXV4-21-&22-H[ A-A24- 9MERS-PSCA Each peptide is a portion of SEQ IDI NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position' plus e ight.
~TRSUBSEQUENCE ISCORE I77 ASVPLLTDL ~84 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXVI-V21 &22-HLA-A24- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight, STRISUBSEQUENCE 7SCORE 1 DLAQWEPVL 4.00n 2 L.sVPLLTDLA 0.180_ _LTDLAQWE 0.017r 7 TDLAQWEPV 0015 4 [PLLTDLAQWV 0o.015 461 LTLQWEP 10.011:1 TABLEXVI-V22-HL11A-A24-9MVERS-:
PSCA
Each peptide is a portion of SEQ I D NO: 8; each start position isM specified, the length of peptide is 9! amino acids, and the end position for each peptide is the start position plus eight.
START [SUBSEQ;UENC SCORE.
I [ASVPLILTHL 8.640 =8 LAQWEPVL 400 9 LAQWEPVL 0.15 i. '_FLLHLAQWE fo4 ~6 fLTHLAQWEP 1-0,1, TABLIEW-V24-HL11A-A24-91VERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9.
amino acids, and the end position for each peptide is the start _position plus eight.
STR SUBSEQU ENCEI SCORE, MVCTVPH 0.750! L-1__LTMY Tv 0.-2 [TABLEXVI-V24 HLA-A24-9MERS- P.SCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[START SSqUECEI SCORE 1 1 fASLTMYVCT _050 IF 7 f VCTPVPHPD F 0076p 8F 7 q TPVPHPiP 0015-i] 2 SLMYVCP 0010 1F7 f Y~vcTPVPI2 ~-IP 0.1 {TALEXI-V5-HLA-A24-9MERS-, Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is9 amino acids, and the end position for each peptide is the start p._osition Plus eight.
STT FIJBSE QUENCE-I7 SCORE, 2 tPT QSS l[0.360 SSID[ ERTDPPA 0. 180 F :&-7TRQISIDT, 0.015 71 S RTPTRQS [0014] l SSIDTDPP 0.010 4 PTRQISSID 001 TABLEXVI-V25&26-HLA-A24 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
START' SUBSEQUENCE, SCORE-1 7 SSSTDPPA 06.120 f TPTRQISSS [6T6 f RQISSSDTD 003 FTABLEXVI-V25&26-HLA-A24- -9MERS-PSCA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight [,SART] SUBSEQUENCE[SCORE _I J. TRQISSDT 100715J S4ISSSDTDPP Io 5 Q1 ISSSDTD 01071 2 PTRQISSSD, 000 1 TABLEXVI-V26-H LA-A24-9M ERS- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight._ 1 JSTART SUBSEQUENCEI SCOREi F7 77G~ T J 0.120 IF I TPTRQIGSS F 0.100 F4 ZFR-QIGSSFJTD [O0030 3 f TfRQIGSS6T_ 0f.01__ Sf8]SDTDPPAD I 6 lGSSDTDPP 40.9010 QI GSD 0f.oio Fi[ 9 S-DTP_DG [P K_ [TAB LEXVI.V27-HLA-A24-9MERS-
P-SCA-JI
AEach peptide is a portion of SEQ 1ID NO: 8; each start position is specified, the length of peptide is 91 amino acids, and the end position for each peptide is the start position plus eight.
START[ IUBSEQU ENCE1 SO~ 27~ RGQALRRAQ 0.036i1 F SRGQALRRA__07i10 TABLEXVII-VI-HLA-A24-
IOMERS-PSCA
WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
[START SUBES E~QUE NC q SCOR:E Ez .JLSNEDCL [12.000o jf108]? ALLPALGLLL 8.640 4V--LLL -LMAGL 7.200K 7.200 98 I-IALQPAAL 6.000 76 j YVGKKTi 6.000 6fIi [L LLMAGLLo6.00 L CNASGAHAL 6.000 LiD[J CLQV-ENCTQL 00 100-6T L.QPAAILAL F 600 59 F LTVSCL w -qT6.000 2- [13O-7 LLQPALL 1 6.000-6 f1-0-1 [QPAAILALL 5.600 11 3 IYSCAQV 4,.000 106 i1ILPAGL1400 F1 TAR 1 A VLL 4.000 7L 1_4.0 F744I[ LGEQCWTARI 11.00 2 FKAVLLALLMA [10 OL FS LQ 0.238 57 G[-LTVISKE7C 0.210 52 RRAVLLTV_ f0.200 F99 7-A-LqPAALA 01-80: iLNCVDDSQDYY: 0.180! i4 ALQPGTALLC 16.180, I~ TLOS1 0 .165 j 74 SQDYYVG KKN [0.54 EI7IL PLqLLW 1 0150 s PCALC1 0.150J ID 7 42 TQLGEQCWTA, 0.150 -TABLEXVII-V!-HL A-A24- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
F S U[qBSEQU ENCE S C~ORE E4C ZLTLGEQCWT1F[0.150 F7[11[G LLQPqTA Rf 0.150 f86 f CDTDL CNAS 1[O.4 4TCTCN [0.12074 [:70 9YDQ Yf 0 .120 39 ENCTQLGEQC 0.2 68 LNCVDDSQIDY 0.120~l 53 .RALLV 0120? 94 ASGAHALQPAl 0,120j 95 SGH QP 0.2 16 UPTALLCYSIO.120 10 ~AALLQP T 1 0.20 4F~7 QCWTARIRAV 0.10 5 hI -VGKNI c F0.1006- I 5 1 LLALLMAGLA Pi0.0 7jQDYYVKKNI---- 1 0.100 i84 ITCCDTDC 0.100 1 L CSH 0.100 65 [6'-cSLNOVDDS L.10o0 103 11AAAILALLPA 0.1007 [7i LLCYSCKAQV 097 46 EQCWTAPJRA 0.10 191 GAHALQPAAA7O0,100 29 QVSNEDCLQV 1 0F,100? f 7 MKAVLLL o 0060 'LK[VLLVIK I .023] 36FLQYENCQLG 4 0O.022 ITABLEXVIIkV1.HLA-A24-__ Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the 'start position plus nine.
3" FVSNE-CLQVE Ij a0221 11[ 94I7 1 GCSLN YDD 122?9i-0 7_ !3,iPSqDqYYVGKK11 .20 1 .1 LLMALP 0.018 L 37R1 DC LQVENCT 11 .0171] 28 FAqvsNECLQI 9:9151 34 37 QVENTLE. 0.01 7 AF LAGAL 005 F748 _CWVTARIRAVG_'j0.014I [7.9-.VGKKNITCCD 14 P 7 PGqTL CYSC 0. 012 1 3 Q(GEQC TR 56T [LMAALALQ-'I0.012 i10 IPAL:GLLLW6 -0.012 F32 NEDCLQVENG{0I01F NASGAHALQP 1f 0,010i~ Y192 tCNA-SGAHALQif 0.010 .80 GKKNITCCDT i ,1 TBEXV11-V4-HLA-A24.
Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the sta' _rt position plus nine.,-i WO 2005/014780 WO 205/04780PCT/1§S2004/017231 iSTART][UBSEQUENCE1ISCORE [159 {A PPDL 1[3.00 13 CFHGAFT [4.000 T317 F[ RPSLRCSL FJ 1 .2q 1[STNFLR-L 8.400 [64 [:GPQASVPL F7,209 =8LI[VPEAHPNASL [6.000 L 5J[ VVPQASVYPLL[6.000 1I-SjLAPSRGQAL j[5.760 [102 i VPPMA 4.800 27 j[MGRPPL[.0 [OSDPASYRL 4,800 14LJ TLNPVLRHLF i 432 [130 GPSN LCC j400 7fro3 ALSRTPTRQI Fi.000 100 APV PHPDPP 0o.900 96 31 ,M-YVgAPVPHP- 0.750 THPAQWEPVL' 0.600 E-1FAHPNASLTM 0.600 126 1PADGPSNPL 0.576I 5T1][SY LWP 0.500 E 3 PAGPMC SR .0.
ii' lRTS§RAVTPTC 0q.280 1 E53LRLW3APLiQPT .240 79 WEPVLVPEA 10. 238 VPiIiiAK7 0.216 66 71FvPQASVPLLT 0.21 ,0 F i] j q S RITACRCS 0200 1831 VLVEAHPA 10.180 F171 f VPLLTHPAQ Ef TABLEXVII-V4-HLA-A24- 10IMERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptidle is the iEstart position plus nine.
FSUBSIPEQUENCE] SCORE~ Is* -F6FG APLQPTLG][0.150 S1586 1 PQiEAFPAHPI 1 0.160 AP[LQPTLGV.V r[ 0150 129 DGPSPCC_[O! 1 iL ATPAGPMVPCS [77i d F 175 VS AGQ 001 I1 F1 _LTMYVCAP [0.1 60 1 PHLS [~A10.140 Y77 VR HLFPQEAP F[ o-i~ 11123.1 DTDPPADGP90120 15 A\LTPTATPA 0.120 20ICATPAGPMPC I0.120I 141GPAFSTLNPV 0. 120., F74J LHPAqkPV0 1[.13 FC6CFHGPAFS: 0.100_1 .,I.SACCSGPA 1 010 V76I HPAQWEPVL'T! 0.100 C7 j LSQVVSVVS0.100 ___CRLPPSLRC_[0.100 I HSACCSGDPA' 0.100 1A8-1 DPASYRLWGAI 0. 10 61 1191_ IGSIDTDPPA 0.100 43 !ACGDASY 0.100 621TGWPQASVI 0.100 F-137]' iCOFHGPAFST 100~l 1-2 j[TSRAVTPTCA F1 ooo 7F7LN [A SLTMYVcAi o 7~ TWARRTSrAV 0.100 TABLEXVIV4-LA-A24- [Each peptide is a portion of SE ID NO: 8; each start position isQ specified, the length of peptide is 10 amino acids, and the end position for each peptidle is the start position plus nine.__ STAR S UBSEQUENCE jCORE 1I 154 _FPQEAFPAH 009 18 ii-TPTCATPGM ][.050 143 AFSLNVLR 0[.050 [117I RQGSITDP 0r .042 RAK---vTPTCATP Jjo:o3oi 1661 iYDLSQ VWSW .21] 1OiRRT 124 [:TDP PADGPSN 61 8~ f155 FPQ-EFPAHP 0.018 I 1,33 NPLqCCFHGP 001 12 .GSIDTDPPADJ 0.018 I1 78 IAQ-WEPVLVPEI 10.017 1 24 AGAP M-PC SR FL -1 .5 I TMY VCAPVP 11 0.015 1 9 PHPDPP 1IiT J.1-40 J.HGPAFSTLNPf0.015 [T7 FPSLRCSL-HSA 1015 [.107 PPM-ALSRTPT 0.0 5157, [17FLNPVLRHLFP 0.015s -TABLEXVII-V1 9-H LA-A24- 1OMERS-FSCA Eahppieis a portion of SEQ Eac NO:t8;each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptidle is the start position plus nine.
ISTARTISUBSEQUE NCE SCORE? 8 f RLFRCL 17.280J 1E I IGMCL I 6.00 4 MP: CSRLLPSL 4.80 WO 2005/014780 WO 205/04780PCT/US2004!017231 TABLEXVI I-VI 9-H LA-A24- 1 0MERS-PSCA.........
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the startposition plus ne.
IITAT SBEUNCE
SCOREJ
IF I[ CRLLP SLR C 000 Li77iJ[ GPMPC§RLLFj1 LLPE- SRCLH F.015 57E PCL LPSLR o 0.01 7 TABLEXVII.V20-HLA-A24- Each peptide is a portion of SEQ ID NO each start position is specified, the length of peptide is 1 amnino acids, and the and position for each peptide is the start position ___igs nine.
Si SUBSEQUENC START E SCORE! 6 P SSRWGA 0-.120 2 C~qCSGDPASSR 0.01 GDPASSRL GIr0.9027' TABLEXVII-V21-HLA-A24-7 !F O1MERS-PSCA Each peptide is a portion of SEQ ID: NO: 8; each start position is specified, the length of peptide is 101j amino acids, and the end position for each peptide is the start position' plus nine.
m~SUBSEQUENC SOR fTABLEXVU=-V21-H-LAA24- 1Each peptide is a portion of SEQ ID NO' 8; each start position is Ispecfied, the length of peptide is 10; amino acids, and the end position for each peptide is the start position IL plus nine.
ST T SUBSEQUENCj
CR
9 TDPAQWEVL 0600O ti [VLD AQWvft, 0q.1507C D07 QkEPVLK![0.100 I [ASVPPLTD1 .0011 TABLEXVII-V21&2HA24 1OMERS-PSCA IEach peptide is a portion of SEQ !jID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the star psition plus nine.
S SUBSEQU ENC E, SCOR E 1_.QSVRLLTDL 4.00j 8-1 TDLQPVq-L 0.600-7 2 ASVPLLT 0.216 F7 FLTDLAQWEPV 0.100 9 D7 [Y VLVZ 0.100 1'D7F-rSVPLLYT-Di--A7 o.01 I LLDEACIW EP cL01P ~IPLLTDLAQWE $[0.002 TABLEXVII-V22-HLA-A24- 10MERS-PSCA_ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position IFplus nine.
SBSEQUENIC
TEAR T E SCOREI l7- THLA Q WEPVJ =0.600 3Rn AVPLLTLA .26f 4 T[ PLHAQ 0.15 I= 10 LAWEPVVP][0.015 :JT=ABLEXVW1V24-HLA-A24- Each peptide is a portion of SEQ ID NO: 8; each start position is 1 specified, the length of peptide is 1C amino acids, and the end position for each peptide is the start position plus nine.
SATISUBSEQUENC
1
SQR
T PVPHPDPPMI -900 I7 YVCTiFV-PH- 0750~ 47 TMYVCTPVPI 0.015 7 YVCTP-VHD 0O.014 T- MY VCTPVPH 00,6107 ATABLEXVII-V25-l-LA-A24-
IOMERS-PSCA
Each peptide is a portion of SEQ ID' NO: 8; each start position is specified, the length of peptide is l0i amino acids, and the end position for each peptide is the start position plus nine. SO~ ISTART' UBEQUN -COE 1 LSRPTRQIS. 0.o120 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXVII-V25-HLA-A24 [Each peptide is a portion of SEQ ID a~NO: 8; each start position is Ispecified, the length of peptide is 10 jamino acids, and the end position fo ahetde is the start position foah ppue nine.
[START SUBSEQUENG
PSCOORE'
ISSIDTP O08 I ljTTQSI r.04 j[ SRTPRQS 001 8F ISSIDTPP ][oj i _q~LPTQSSIDT o oi if TABLE HLA..A24- Each peptidle is a portion of SEQ ID, NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptidle is the start position .plus nine., SO BSEQUENC I START'
SCORE
E.
i_ [RIP TTRQISSS] 0.300,, 7 PI RQSSTP 6 if72JTP-TRqISSSD 5014i 8 7 '[-SSDTDPAi 0012.
:F 6 FQISSDTDPPF 1 0.010 3~7 TRQISSDT 0.010 1 4 TRQISTD 0.002 S TABLEXVII-V 27-HLA-A24f ach peptide is a portion of SEQ ID! NO: 8; each start position is specified, the length of peptide is 10! amino acids, and the end position for each peptide is the start position' plus nine.
ISTART SU EQUENG SCOREj
E
I PSRGALRRA 0.010 TABLEXVII-V27-HLA-A24- 1OMVERS-PSCA
J
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
[START SUSEQUENC[SC0RE1, ITABLEXVIII-Vl -HLA-B7-9MERS- Each pepile is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 Iamino acids, and the end position for each peptidle is the start position, plus eight.
START SUBSEQUENC SCORE~
E
IF -i~GV 6 10 ioFF QPAML A LI I[ 8000 r-60 IrV IS KGCS L 0ooo E0 13 DLLPGTAlfl&000'q 72 AQVyNEDCL 1200 105 iFAI LLPA ,L1200 V108 GLLPALGL 1 12,000] 99 A AL QPAAAL_j 12.0 j 4 I ALLA Lj 200 F ALQPG TAL [1-2.000 92 f NASGAHAL [4.000 LVENTQL4. 000 83 NTCCPT DIL -4.000 D o6 FLL PALLL7 F4.o 5: 7 E I L AMGL F4.00 F 115 'IiE L PiI 4.000 98 Jf HALQPAA 1.80 I64.ThVLLTVI 1.200'T~ '51 If A RRVL .0 TABLEXV111VI-HILA-B7-9MVERS- Each peptidle is a portion of SEQ ID NO: 2; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start position plus eight, jSTRT SUBSEQUENOI SCOREI R IRAVGLLT j. 1 000 104 AALALLA L[90067 78 [i K, Y NTC 0 K16 QPGTALLCY_ 0.400 11 LPLLLW,0.400 F 0O.400 55 AVGLLTVIS 0.300 H GAALPAA 0.300 '11 AGLLQPG 0300 0, LALAA 0300 30 4 VSNEDC 0.200 QCWTARiRA j, 0 150 -34 [LQ NC [00 18, GTALLCYC 00 [95 SGHALQA]oio I7![VGKKNITCCO 100 I 40 [T~TQLG QC 1 -io i 43 r QLG-EQCWTA. 0o.1007 I84 _ITC(CDTDLC 0100K F 91 FLC(;NA SGAH 0.100 42 6FiLGEq6cW i 010 0 .78s8 LLTVISGC 0.0 29-1_QVS NECQ 000 FT 71 F~DYYVGKKNI f .040 94 I ASGHALP F. WO 2005/014780 WO 205/04780PCT/1§S2004/017231 FTABLEXVIII-V1-HLA-87-9MERS- PS-CA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plseight.
S aTART nUBSEQUENCI
SORE
112- f ALGqL LWGP I003 19 ALSC 0.3 if 861[7 CCDTLN f .0 30 69 NCVDDSQDY[002 63 IFSKGCSLNOV CEX TQLC [0 F. 020 61 4 [VISPKGCSqLN IF 42o7 I [CSLNCVDDST
W
53 I) V GLLTV F C.~9 48JWTARIRAV_1F 0.0 20-, 24 SCAQVSN 0.20 49 WTAIRVNE 0.05 4 [KGOSLNCVD_ 0U10 165 LGSLNGD -1k91,9 FI 113 E GLWGPG '0.0610 .1 CLQVENCTQCj .01O7 [7 SLNCVD DSQ 0.010 8 KKNITCCDT .[Ti.i 1 94ENCTQLGEQ I .oioj 114 LLLGPGQ 010 73 DQY K 10.010 [TABLEXVIII-VI-HLA-B7-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position ifor each peptide is the start position plus eight. START1jSUBSEQUENC j 9 fLMAGLALQP 0f .010 F ILC1AVSbnEK [0.010 106 jFIALAG~ 0.010 If78 fT NASGA i 0.010 if 77 YYGKNT if0.010 EL I VLLLLMAGif01 6, v1 VGLLVIS 697p.o I TABLEXV11114-H LA-B37-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9' amino acids, and the end positionI for each peptide is the start ____position plus eight.
[START! SUBSEQUE..NCESCR I5 32 LPSLRCSLi 20.000 F11PAHPIYDL F879obbf 667 VAPLL 18.000 5 6 PWEPL 1800 VVQ.VP Ill~~ _STPTRQS 670007, 8 WARRTSRAV if6,000 163 7PYDLSQV [40907 F146 TLNPVLRL 4.0 1 51 J7SYRWGAPL J.00 55 E7WG6PLPjjp7 4.00 130~ GPSNPLCCC 300 17 5SPAPSRGQA 3.000 b 102 4 VPHPDPPM 2.000 ITABLEXVlII-V4-HLA-B7-9MERS- IfEach peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 Iamino acids, and the end position Sfor each peptide is the start po,__Rston plusigt STARk- SUBSEQUENCE SORE 106 DPPM ALSR .0 19 MTATPA TM .500- L177 P SRQAL I 1.200.
L46_ [GDP ASY RL E01.] VFB3 jAFSTLNPVL 35 RCSLHS§A 2-0-o-, [101 PTPDPM ]I[0.750J L LGMPCSRLP I .0 22 TPFAGPMlPC 0f._600 94 7,LTM YVCAY V [q 06 0 178 APSRGQALR 0.60 1J0 A PVY PP 0.6 00- 1[27 f ADGP SNPL f0.5409 4f_LvPEAHPNA7.q LOO i170 f QVWS~VPA 0o.500 _70fsVPLLTHPA 050 9 ;f S ARRAV f 0.450 0.00 F 27 [mP~C PPS 0.400 1-3-9- 7FHPAFTL 1 0.400 88 A PNASLTM I OTO 42 6ACGDP 15300 V48 DPASYRLWG_*[o.
21 TPAGPMPC 1 0.300 C3 LGVVPQA6 T.0 92:_ AS LTMYvcA- 0.300 4j 87 1 EAHPNAS L LO.300; 91_F_NA SLTMYVC 1 0.300 14 FNPVLRHLFP_ 0.FO200 167 LSQVSVV f 0200q i6 8[EPLVPEAH_ F 6.200oi WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXVIII-V4-HLA-B7-9MERS- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptide is the start osition plus eight.
STRTI [S UBSEqUE NO E S COR E Q PTLGVV
PQ
_133 ifNPCCFHG 17 ]f FTPTCATPA200]-?o F71 ]f THPQ ,700 11 4 PA-FSTLN _0.0 tlj HDPPMALS 0,180]- 29 CSqRLPSR 01j 7 VPTAT .150 129 [I DGPS-NP LC q[ 0100 1 I 6 TTATPA !10.100 1-20 GSIDT DPA_ 6 01001 L RTWARR T 0 IH571[ LRHL FPQEI P.1 o01 135 IILCCCFHGPA 0.1006- 1 MTHRTTTVVA I010 [i731 J7YVySPAPSRT 05 [977Y-VCAPVPHP .90 75~ 1- 43 1 ACOSGORAS--0 -6 0 F EAFP AHPIY_ ].6 86E HNALi 0,060 103 4HPDPPMAL 06 7 7: AQ WE PvLV 006 7042 AF§TLNPV 0.060 15I7 -QEAFPAH PIf 0,060 174 [WSPAPSGL.5 99 1CA -PVHD [0.045 78,1 QWEPVLV 0.45" AS-YRLWA 0.030 TABLEXVIII-V4-HLA-B7-9MERS-
PSCA
Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of paptide is 91 amino acids, and the end position for each peptide is the start p ositioin plus eight. I 12F ADGPS NPL1C EO .03OJ 56 PLQPTLq 0."(30] 109: E MAl RTPTR 0.00C 69 SVP LTH P L .o3 oI 57 TTVARRTS 0. 03-0 20 j CTPAGPMP L.3 =147 fLNP VLRHLFI.03-1 :F13 j CFHGPEAFS rL0.20 126 P G S [I o.o20 if 1 RPPLRCS H 002 [I TA BLEXVIII4116:WCA--BM-MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is Ispecified, the length of peptide is 9 amino acids, and the and position for each peptide is the start position plus eight.
I[START SUBSEQUENCI SCORE-.l I G PCSRLL 1240.000: 8 4LLPSLRCSL,600 3 W MPCSRLLPS 60.400 f-9 F'CPL cs H'F0.20 5 7CSRLPR [0.100 7j RLLPSLRCS if0.020 1 K6[ 1SRLLIPSILRC [0o.05I F-2IF PMPCSRLplP~0b Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight______ sr~r SUSEQUEN COR 8. S S RLW GAPLJ 00 1 3- FSGDPA~SRL 1 200s L6PsSR [[030 6 7 PPASSRLW !000 9 SRLW GAPL9 [01F ~TABL XVIII-211-H11A-B37-9MAERS- I LE PSCA fEach peptide is a poto fSQID NO: 8; each start position is Sspecified, the length of peplidle is 9 Iamino acids, and the end position Ifor each peptide is the start positionf plus eight.
;START SUSQE SCORE11 I0.000' 3 'PLLTDPA 0O.500 4 PLL TDPAQ F 020 i qXSVIPLILTID 40.0451 ASVP-LLTP f0.3 T 8 -T -DPAQWVEPVF 0.020 8 LLTDPAQWE 70.015 7 LTDPAQWEP 0.003J.
C[PLTDPAQW[ .0 TABLEXViIIkV21 &22.HL-- 9MERS-PSCA Eac petde is a portion of SEQ I D NO each start position is specified the length of peptide is 9 amino acids, and the end position for each peptide is the start position i plus eight. WO 2005/014780 WO 205/04780PCT/1§S2004/017231 It SUBSEQUENC E START _E SCE KCI1 AV-PLLTDL j 12.OOO1 F81? DLAQWEPV L [400 2Lj S PLTDLA, [0.500 3 VPLLTD-LAQ 0.20 7 [TDLAQWE V 0 O~ LLT DLAQWE 0.010 4 PTDLAQW f 0.002:] TABLEXVIII-V22-HLA-B7-9MERS-I Each peptide is a portion of SEQ ID' NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position: p___lus eight.
'[START SUBSEQUENCISCOR [1 {ASVPLLTHL [200 F 8 ifHLAQ-WE P vL IF .6 6o K 7EL LAQWEP-VLV EL.0 2 FSVPLLTHLA if0.5607 1--7 []tHLAQdWPFVJ[O.020 :F 5 F THLAQwE Vo-I6 1 i -Z 4 ][PLLTHLAQW 0.02j STABLEXVIII-V24-HLA-B7-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
STAT'IUBEQUENCEFSCORE
3 LTMYVCTPV ~[0.600 liiiASLMYVC ~[0.300 I77 YVCTFPPP 0.1075 0.015 LLL 71 FTMYV-C-TvP7 J CTPVPHP 0.010 I TABLEXVI ll-V24-HLA-B7-9MERS-
PSCA
Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position L- plus eight.
[START1 SUBSEQUENCEi SCORE SLTMYVCTP 0.010 1[ 5, MY VCTP3VPH i 0001 TABLEXVI II-V25HLA-B7-9MERS-
PSCA-
!Each eptile is a portion of SEQ ID~ NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[TARTSUBSEQUENCE
SCOR
J[ TPTRQISSl 1[ 8000 79 1--SISITDPIPA [oioo 1--2 jRTPTRQISS 0.020 8 ISSIDTDPP 1 Fooi 7 f ISS1DTDP 0.01 4 1 T PTQidI[ 0.010w I RQISSIDTD_ 0.01 5F97 -TRQISSIDT 'j0.010_ 1 S- L§RTPTRQIS7 oAo3 TABL EXVIII-V2526-HlA- .9MERS-PSCA Each peptidle is a portion of SEQ* I DI NO: 8; each start position is specified, the length cf peptidle is 9 amlno acids, and the end position for each peptidle is the start position;1 plus eight.
START, SUSQEOSCOREi
E
F"1 ,,:TPTRQISSSI 0.400 LF7iilSSST-DPP A 110.100.
ii if QSSSDDP f0.010 6- ISSDTDPP 001 3 TRQISSSD 1 0.010 1 4 QSSSDTD 11 0.010 [TABLEXVIII-V26-HLA-B7-9MERS- L PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 91 amino acids, and the end positionI for each peptide is the start position pluseight.
JSAT[SUBSEQUENC SCOREi E 1 PTRIGS ~f0.4001 f q F Q SS TDP f 0.010 6 qITDP 6 3 fTRG-SST 0.010 If2 PTqIG(S-SDT j c001io K_8 SSTD1PP f 0.0101 9 [S§-DT-D-PPAD 0L.00321 ITABLEXVIII-V27-HLA-B7-9MERS- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids, 1and the end position for each peptidle is the start position plus eight.
I TR SUBSEQUENCE SCORE f~7RGQALRRAQ 1 SGQALRRA 0.010 TABLEXIXV1 HLA-B7-10MERS- PSCA IEach peptide is aprIon of SEQ ID I NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
[F-50 fkTA iRAVGLL 1,F20.000 Q2PAAAILALL 19104 AAILALLPAL 36.000 18 ALLPALGLLL f2oj [TOi- LALLPALGLL__ 12.00 98fHALQPMAAIL 6JLALLMAGLAL 112,00076 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 fTABLEXIX-V1-lA-7-1 OMERS- Each peptide is a portion of SEQ IDI NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
[ITRT SUBSEQU-ENCE SCORE! 27 K..AQVSNEDCL[1.0 [F1-37[ LALQPGTALL [12.000 KiLILALLPALGL [6.0 [F 35 CqVENCTQL [4.000 If 91 LNASGAHA 400 f 114JI GLLLWGPGQL 4 .000 IF ijj[ __.LQPAAAILAL !F4.000_ [i iWTARIRAVGL-[ 4000 7LVLLALLMAGL [4.000 fi7 KITCCDTDL [4.000 52 RIRAVGLLTV 0 00-i 9 QSNEDCLQV 1.000 [103' AAILALLPA 40.900 8 YVKNI 1 0.500 16__1I QPGTAL LCYS [0.400 470 CVDSQDYYV .0 IFi7, fAGLALQPGTA [0.3 00 k[ LQPAaldLA 0 .300 M~O [A2IkLLQPG [0.300 i[ 7 IFAHALQPAJA 4 0.300 C9[A ,SqHALQPA I0.300 1 -2 1 KAVLLALLMA 0.300 if 1 ITAD LOY(SOKA 0.300 [14 [ALQPGTALLc 0-T.300 I62 f IK SLV 0.200 i110 LPALGLLLWG 020 0 1- LLCYSCKAQV 0.200T 47j4 Q6WARIRAV 10,200 97 AHALPAAAI 0.180 3 AVLLALLMAG 70150 46 ECIWT.A.RI.R.A 0.150 1 7[AVGLLTVISK i 0.150 i 44._1 .LGECVTARI 0.120 "I7 I CTQLGQCT 00 TABLEXIX-VI-HLA-B7-1 OMERS- PSCA.. Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
=START SUBSEQUENCE [SCR~ F670 TVISKGCSLN 0.100 I GLLTV[KGC 0.10 F~j VISKGCSLNC 0.100 j__LLALLMGL 10.100 9j LCLNASGAHA ji_0.100J 42 7 I _TQLGEQCWTA. 0_.1009 .39 7~NOQLGEQ 010 ]9 J SGAHALQPPA ]1-F0.10 83 1NITCDTDLCIj 0.-100 F- 54J RAVGLLTVIS F0.060 Li-5C~ L-QYYVGKKNI [0.040 F 3FiNI GLTV[0.040 7FE61 l[ ARIRAVGLLT 40.030 1o5 Y-iILLAL [0.030 [112 ALGLIlLWGP G_ 0F.030 8 ][0.039_ t 81AQVSNEDCLQ 40.030 93 [NASGAHALQ Fo .0306 F204 ALLCYSCKAQ 40.030 7 tALLMAGLALQ 40.030 f884l DTDLCNASG7 40.030 [79 NC'/DDSQDYY 0.020 58 LLISGSj.
65[GCSLNCVDDS i .2 68 jLNhCVDDSQD 0.020 I40 I[ NCTQLGEQCW 0.0a20 LLPALGLLLW 40.020~ 48 ITGDTDLCN J0.020 2 LOYSOKAQVSJ 0.020 i [LQPGTALLCY 40.020 137 1LVENOTqLGE .15i j- b CLQVENCTQ 0.01 ECLQVENCT--I 10,010-1 25 CKAQVSNED [0.010 175 TABLEXIX-Vi -HLA-B7-1 OMERS-1 C A Each peptide is a portion of SEQ ID1 NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start2positionmpus nine, START]_SUBSEQUENCE SCORE VLGKKNITCCD f0.010 It 64 kSNCVDD 1f 73__ DSQIJYYVGKK 000 26 CKAQVSNEDC-1 ii_.0 _92 CNSGAH:ALQ] 0.010 80 GKKNITCCDTJ 0. 01 0 76 DYYVGKKNIT 0.010 77 YY__VGKKNITC 0F.010 iF -30,1 VSNED=CLQVE Fi0.1 24 ]YSCKAQVSNE Fwoolo 17f PGTALL-CYS-C 0.010 F I LMAGLALQPG ,~i 56 11 VGLLTVISKG IF 0.01-0 36 I LQVENCTQLG 9 O IlO [7F LGEQCWTAR 07.010_I IF 113 LGLLLWGPGQ 0.010T 6 GTLCNASK 0.010 1['7 1 SQDYYYGKKN 110.006 TALXX4-HL11A-7-0MER-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino iacids, and the end position for each peptide is the start position plus nine.
START: SUBSEQUENO 4SCORE! 102§ VPHPDPPMAL 112000 100 APVPHPPPf 190-900 -27 4MPCSRLPPSL 176 FSPAPSRGQAL 81 64 1GVVPQASVPLE2.0 65 VVPQASVPLL WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXIX-V4-HLA-B37-1 OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each ppide is the start posifion plus nline.
START SUBSEQUNCESCRE, jj ASYRL-WGAPL 1200 APLQPTLGVV 20001 K RLPLRSL 6.000 L.IIWARRTSRAVT 140 14 [GAFSTLNP 100 L 94HPNASLTMYV [4.000 .2-61 HPAQWEPVLV[40 jTLNPVLRHL I4000C 8711 4EAHPNASLTM [3.0001 .LO i QPTLGVVPQA f 2000 {66 [VFQASVPLLT f2 000 48~i PASRLwGA F2.000 19 AFPAHPIY)L[ 1.800i 23 PAGPMPCSRLFT .8oo 110 ALRTPTRQI 1.800 AVTPTCATPA 1.500~ K fj CSRLPPSLRCf150 F1 142 !I PAFSTLNPVL [120 SL-CHSAC fi.o0 Y LRHLFPQEA 1.00 107_i PPMALSRTPT 0.900 PMCSLPP 0.900 ['GAPLQPTLGV 0.900 163 7 HPIYDLSQVWV 0.0 F7 1 RTPTRQIGSI 00 TPAQWEPVL' 0.400 130V~ GPSNPLCCCF [10T400 flajCFHGPAFSTL 0.400 71 I VPLTHPAW 4 400_j 81 EVLVPEAHD 'j0.300._j 4TABLEXIX-V4-HLA-B7-1 OMERS-
PSCA
Each peptide Is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptidle is the start position plus nine.
[START-i[SUBSEQUENCE ISCORE F6 2,1 TLGPQ-ASV 0.300 20 CATPAGPMPCK If0.300 9 j[NASLTMYVCA If0.300 [69 4 _ASVPLLTHPA [0.300, lj-5:I DPPADGPSNr- Pf-- I 0.20 3 [LPPSLROSLH 0.200 -2 IfTPAGPMPCSR if 0.200 1 IfT ."_PPMALSRTP-ILO.2001 155 FPQEAFPAHP '0-.00( L 11 TPTCATPAGP 1[1200 F1-33 7 NPLCCCFHG;P 0.20 0 I 93 LTMYVCA-PV, 0.;.200 !114F TPTRQIGSID___ 500 I 44LTHPAQWEPV 1[0.200 53 RWGAPLQPT j 0.150 iF57 LSRTPTRQIG 0.159 129, fDPSNPLCCC if0.150 Ki' J VSPAPSRGQA 0.150' isiPTcATPAGPM ~[0.150 179 7 f PSRGQ.ALRRA If0. 10 0 1 _1 73 [1 HLFPQEAFPA FO-1 i 11 RTSRAVT PTC' 0.100 6 j TTWARRTSRA :yo' [f 83 [,VLVPEAHPNA [.0 F84 7LAPN i -0.100 [19F _1sQ'/WSSPA j0.100 ,1Th IfPTRQIGSIDT 40.100 41 HSCCSGDPA][ o1o 21C 1 APAGPM-PCIS4 0-090 7 vsPAPSRGQ 0.075 F 7 F'H6PPmALsR~ 0060 W 162 AHPIYDLSQV _.960 :7CCSGDPAS 0.060 176 TABLEXIX-V4-HLA-B7-IOMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
S~TARTI SUBSEQUENCE Is~o EI -43 ACCSG1DPASY-10.06 710 PVPHPDPPMA 70: SVPLLTHPAQ 0.5 0 I91YVCAP VPHlPD ,0.050 _f2 f_AGPMPCSRLP :f 0.030] I[!46 IfTLNPVI-IL .3 4 -R7f LTMYVOAPVP7,[ o.030 L2_ jjf ASLTMYCAPj 4_.073_ IF 128 if ADGPSNPLCCI .3 Q6 V' Th PLLTHP pQ V 9 ARSRA 09.030 78AQWEPVLVPE 0.030 14 RF PCT 0.030 99 CAP VPHPDPP 003 10 I NALSRT TRQ 0L..030 44 RTTTWARRTS !fA03 ,15 '8 FPAHRYD 0.030 if 16 YLSQV'iSVV 0.2 TABLEXIX-i 9-LA-B37-1 MERS-I
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified,: the length of peptide is 10 amino acids, and the end position for each peptide is the star position plus nine.,i START. SUBSQUEC SCORE 4 8C.000 1 1-1 AGPMPCSRLL. 12.000 8 R-7FLLPSLRSL[ 6.0007 I -ICSRLLPSL RC 1.-500 2[-I GPMPCSRLLP. .01 I-'17 0 Sl- LSRC S-LHS7 0.400 9 fLLPSLRCSLH'011 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLE19-HL11A-B7 IMERS- PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino I [acids, and the end position for each peptide is the start position plus nine.
START SUSQECE I SCORE PPCSRLLPS ]0.002.
7 SR1'SLCS0.002 TABLEXIX-V20-HLA-B7-IOMERS-
PSCA
Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
START SULBSEQ9UENq CEIsCORE1 I 8 1ASSRLWGAPIL~ 12-- .000 1[ 1CSGDP -ASSRL]F _j 00 6DPASSRLW GA 2-1.000 0SLGP .1100 I [ACOSGOPASS I 0.060 I2 CCSGDPASSR 01 4 [SGDPASSRLW 0.006 PA 1,±SSRLWGAP 0,003 L' 10- SRLWGAPLQPU 0:0J ~TABLEXX-1-i..HA-B37-11OMERS- SEach peptide is a portion of SEQ ID NO: 8; each start position is specified,! the length of peptide is 10 amino acids, and the end position for each pept ida is, the start position plus nine.
STRT SUBSEQUENCE lSCOR DAQWPVL l[4.000 'K ITD PAQWEPV 0.400 4AVPLLTDA 03400 4 SVLLTPAQ 0.050 L 77QASVPLL DP FC030 LLTDPAQWEP I 0010- ITABLEXIX-21-HL11A-B37-IOMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
LST ART SUBSEQUENCE ISCORE, F PLLTDPAQWE 0.002 1 QAVPLLTD 0. 002 [TABLEXIX-V21 &22-HLA-B7-1 OMERS- Each peptidle is a portion of SEQ ID I NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
START [SUBSEUEC SCORE: 1 4QASVPLLTDL 12.1000~ 87FTDLAQWVL 7 0.400 1 F 1 VPLLTDLQW 0.400] 2 ASVPLLTDLA 0.300 9T DLAQWEPVLV[ 0.200 3F -"ISVPLLTDLAQ 0.050 6 ~LTDLQWEP 1 0.010 ____PLLTDLAQW 0.001 ABLEXIX-V22-H11A-B37-11OMERS- Each peptide is a portion of SEQ ID1 NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptide is the start position plus nine.
FTR F SEQUENCE FSCOREi I QASPLLTHL 1200 [777 THL6Q'WEPVL-_[ 0.40 1 -VPLLTHLAQW If 400 2F ASVPLLTHLA f_.300 i FT LTHLAQWEPV F0.200 17 9 f- H.LAQWE.PV.LV' 0T.200 L% 7IVPLLTHLAQ I 1.5) 6 LTHLAQWPli61~ EI F D -4itH~~i~o 177 I TABLEXIM-24-1-11A-137-1 OMERS- -PSCA 11 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptidle is the start position plus nine.
STARTJ UBSEQUENCE SCOqRE 1 0111 TPVPHPDPPM 3F0.000 1 ALTMYVCT 0.a300 IF[SLTMYVCTPV_0.0 'F i If YVCTPVPHPD IL 909 7[ ASLTMYVCTP 0~3o0 4 IfLTMYVCTPVP II0030 IY- qFCPVPPDPP 0.010, s If TMYVCTPVPH i[ 0.010 I~ TABLEXIX-V25-HLA-B7-1 OMERS-
PSCA__
Each peptide is a portion of SEQ ID INO: 8; each start position is specified, tihe length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
STARTj SUBSEQUENCE F CO F _TPTRQI SSD 9 ISSDTDP 0.10 8 QISSIDTOPP 0.0101 f2 CRTTR Q IS 0.002 Xq 6 rT-RQI-sSI 0.001-T, fITABLEXIX-V25&2-HLA-B7-1 OMERS-]
PSCA
Each peptide is a portion of SEQ IDI NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each pePti de is the start position plus nine.j START1 SUBSEQUENCE SCORqEl WO 2005/014780 WO 205/04780PCT/1§S2004/017231 rTABLEXIX-V25&26-HLA-B7-IOMERS-,
PSCA
Eahppieis a portion of SEQ IDI NO p;eptidstart Position is specified, th eghof peptide is 10 amino acis, and the end position for each pe ptidei the start position plus nine.j [fsmART 3[ SUBSQEC SCORE I IF-_ IS.lSSDTDPPA [010 K 39L= iRQlSSST Ij[ 0.0 1 [RTPTRQISSS [0.020 K Ii RISSSDTDP TR1--QSSSDTD 0. 000 ITAIBLEXIX-V26-HLA-B7-1 0ES
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified,, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
ISAS1UBSE'QUEC E FSCO0RE I2 ITPTRQlGSSD 0.200 PTRQIGSSDT 0.100 7. jISSDTPPfA 0.100 F_9 SSTDP ADG .0 7 SDTDPADG~ P 002 ITABLEXIX-V27-HLA-B7-1 OMERS-
PSCA
Each peptidle is a portion of SEQ ID INO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each Ipeptide is the start position plus nine.I
[START
1 J SUBSEQUENCEj SCqR El 1 PSRGQALRRA 0,109 F77 j[7G9A RAQ 0.00 TABLEX X-V1 -HLA-B3501 -9MERS- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide Iis the start posiion plus eight.
=SA U[IBSEQUENCE,[SCOqR:E F165 QPGTALLC 40.000 2 KAvLLALLm 1 12.000i 7if. i 1_ Lf!LPAGLLLW 1000 10j7 LALLPALGL 9 .000 t I SNEDLQV 3.000 ji71 LQPTL 3.000 f_ RVGLLTVI 1 2.400_ 38 IL_LQVENCTqL 2[.000, 621 I IKGCSLN-C- 1. 676 F706 E 7CVYDDs-QDYY__TLi.2oU 7fALQ 100 60~ TVSGS 100 0 108 if LPALGLL 105 ifAILALLPAL if i.0o LLLWGPGQ LJ 1.00 83_I1 NlTqCDTDL,, 1.010_9 I_ 1..,ALQPGTALL_ 1E6000 28 17 AQGVSNEDCL 1I000 .F 92 1 NASGAHAL I .0 F' 77 sf LLALLMAGL FIlOQO 19 LLPALGLLL 1.001 7 fALLMAGLAL 1.0 52 FRRAVGLLT Fs~o.
[77 FKAQVSNEDC I06 2 IfYSCKAQVSN f!& 66 CSNCDE 0.500~ jVGKKNITCC 1 00_I [14]AAIALLPA Il0.00 iF 102 11.PAAXALL L O7 96 If GAAqPAA 'F0.300j IF--6] LALLMAGLA--I 10.3 0 0 178 TABLEXX-Vl-HLA-.B3501-9MERS- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids: and the end position for each peptide is the start position plus eight.
START B SBEUNCE [SCREI if _85BS jf TCCDTDLCN__ .0 If_72 LCYSCKAQV Nf0.0 4 fTQLGEQCWT 0.1_0 84 lCCDTDLC_ 015 12ifGLQPGTA '[01 00 1 7 SwDrVqk 'f-9 01001 L71 .i[QCWTARIRA][070 If 18 If GTLLCYS-C 0 .100_ i EL _VISKGCSLN Ifoi 15 _LQPGTALLC 010 AVG7yLLTVIS1:160 3 AVLLAlLMA7'0.100J 51_7[ IRAVLLI1 0.10, 95 SGAHAQPA, 0.Ioq 58 LLTVISKGC [011 40 N L QC3 0.10 94_IASGAHALPQ 8 !CCDTDLCNA 0045 4_4GEQCWTARI .4 257 SCAQSNE ,F0.03% 19j I TAPLLOYSOCK..' F,93 NASGAHALQ J,0.30 103 AALP 0.030 MAGLALQPG [73 64 ~fKGCSLNCVD [0020 63l SKGO:,SLN CV f0.020 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Each; peptide is a portion of SEQ ID NO ;each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide L is the sta rt ion plus eight.
S TARTiLSU:BEQ NC SCORE F-8-27 KNITCCDTD 02 536j .IRAVGLL-TV 0 02 81 .MIf TCCDT 0.020 4 fCWTARRAV 0,0~20: If 2 ffQVSNDCL jf0,015 L6 _fLNCVDDSQDI 0.015 CLQVNCTQ 0,015 DLCNASGAH 0.010 EDCLQVENC 0f joi If2 fLLCYSCKAQ Af 0.0i0 I -23 IFCYSCKAQVS [oUoi 112 ALGLLLWGP 0.1 1 ifPGTALLCYS f!i-o* 56 :1_VGLLTVISK 1-.61-7 49 IfWTARIRA VG q .1 1FT MAGLALQP 0.010 3 N C ENTQL G EQ 0.0-10- 6F GCSLNCVDD 0.1 IF977' AHALQPAAA 10.010 L106 ILALLPALG ;[0.010, 8 LLGAQ J, 0.010 I46 QCTARIR 0.010 !FTDLCNASGA 0.1 j TABLEXX-V4-HLA-B3501-9MERS-
PSCA_
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 9 amino acids,' and the end position for each peptide the start position plus eight.
START jSUBSEQUENCE CORE TBLEXX-V4-HLA-B3501-9MERS-
PSCA
Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids,~ and the end position for each peptide ithestart position plus eight.
SUBSEQUNESOE
HPNSLTMY40.000 1 FT67 _-IPAQWEPVl nno 321LPPSLRCSL 2cL 0001 160 YD 2.0..0 0.0 !LC-ii VPQASVPLL 20.000 114_j TTQGS 8~.000~ 116 fHPIYDLSQV _6.000_ 18[EAFPRAHPIY i .0 I 111 LRTPT QI Qq600 57 APLQPTLG 'V IFZ4.000 1 25 DPPADGPSI'J 3.00 102 VP YHPDPPMA r j 00-0 F4 i IccsGDPASY 3.000,,, 130 6 GPSNPLCC 2,000 16 SPAP§RQA-' 2.000 19 TCATPAGP 1 F2.00 V1OW6 7 E7.PPMAL-sRT 2.0-00-- 8 iF A-RRTSRA/I 11.800 12 TSRVTPT ff1.500 147 _7UJV LRHLF 1. 0001 -146_,TNV~H .0 6 VVQASVPL 1.0 120 7 GS D T D PP rf Oq67 ,F55 WAPLQPTL FP100 1%24 E)PM PRL J[T.00 F136 IF CCFHGPAF, 1F-.000 I fHPDPPMALS 60 85 4 PEAHPNAS rQ.6900' f131-11 PSNPLCCCF[ 0.500 [38 1 FCLHSACCS 0.5s67 172WSSPAPS .500J I6 7 LLSQWS -VVS 0.50 179 ifTABLEXX-V4-HLA=B301-9MVERS7I SEach peptide is a portion ofSQI NO: 8;ectrt position is spcified A the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eit.
SARTj SUBSEQUENCE ISOREJ f155-J! FPQEAFPAH i .0 3 j[SLRCSLHSA If0,300 9 ffNASLTMYVC 0.3001 if EAHPNAST 0 17 PAPSRGQAL 00 46 SGDPASYRL 00 SACOGODPA 030 51 If LWGAPL if0,0 EF12l RHLFPQEAF if0.200 71 [VPLLTHPAQ f '0.200 11 fRPRQGS 0200 [Ti TSRAVTPT .200 1 3 NPLCCCFHG 0200 167 Lf DLQvwsVVy f .oq ~fPI EPVLVP EAH I 25 GPMCSRLP 0.200 E178 FAPSR GqALR jF12-00 60IQPTLGIVVPQ 10.200 88 4 HPNASTM 4 .200 1 IL 1--f LTmyvCAPV If .2o If-qq FT 47 PHPDPP '4 0.200 17F- fTTCATPAG .02007 PPT ~fhA Y-LWG If L PVPPDPPM [0.200 37 1 IRCSLHSACCF_6 0.20u 4 fRTTTWARRT_' 0.200 Fg 7 LVYPEHPA F6 2 0 j 141 GPFS 9P,2061 F- SRLT7 PPSLR I f6T---7, 83 jvLVPEAHPN 0.1507 I21 IATPAGP-MPC 0.100' 143f AFSTLNPFEL 0.100 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXX-V4-HLA-B3501 -9MERS- Each peptidle Is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
TART SUBSEQUENCE] SCORE;I [1357 C CCFGA IT 0 Li 39 I FHGPAFSTL_ I 0.10 fTTTWARRTS. 0 .100 f 6 IYDILSQV Jf.100 F 2 4TLGW PQAS 0.100 170 QVWVVSPA I0.100-j [D2 GPSNPL~CC 0.100~ 137 -'-CCFHGPAFS 0.100, 140 HGPAFSTLN 0 0.100~ IF 127 4 PAIDGPSN PL 0.090 l[--7jF PAQWEPVLV 0.090- 2 i PAFSTLNPV F1 0060, 169 ASVPLILTH -P 05 9i4.F FTLNPLR 0.050 -72 7 PLL(THPAQW 0 050 I fVSPAPSRQ [0.050 47 [GIDPASYRLW- W005 -344 PSLRCSLHS F0.050 [AYRLWGAP 0.oo50 1I.FHSACSGDP 5 57 QEAFPAHPI. 0.040I [126 I FPADGPSN__0.040 L _10 VLRHLFPQE 0.030 4TABLEXX-V1 9-HLA-B3501-9ES I. PSCA Each petide is a portion of SEQ ID NO: 8; each start position is specified,' the length of peptidle is 9 amino acids, and the end position for each peptide is the start position plus eight.
STARTI SUBSEQULENCETS-COREJ LLSLRSL f 1000 -7F] RLLPSLRCS__ 0.200_1 Fj ]7 CSRLLPSLR _1_0.150_ 4 P__CSRLLS .0 ~F 7 SRLLPSLRC 0.010 1L1] PMPCSRLLP 000 STABLEXX-V20-HLA-B3501-9MERS-
PSCA
Each petdl is a portion of SEQ ID NO:8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidleI is the start position plus eight. STAT ~SUBEQUNCESCORE: 3GDPASSRL 0.3001 -5 1F DPASSRLWVG ~[0.200 12 CSGDPASS 0.10 F4 [.CGDPASSRW 0.100_ ASSRLWGAP F0.0 9 L SRLWVGAPLQ_ 0.001 TABLEXX-V21-HLA-B3501 -9MERS-
PSCA_
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
S, ATISUBSEQUENCE_SCORE' 1 9 fDPAQEFVL 20.000 14 IIVPLDPAQ 0.200 PLLTDPAQW F0.075 21 IAVPLLTDP 1 .5 ~7 4QASVPLLTD 'F0.030 6F-- -7 ILLTDPAQWE_110,0201 TD PAQwEPV 0.020 7 LTDPAQWEP i .0 rTABLEXX-V21&22-HLA-B3501-1 'F9MERS-PSCA____ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, thelength of peptidle is 9 amino acids, and the end position for each peptide is the start position plus eight.
fiSTART ISUBSEQUENCE iSCORE F 1 f ASVLLTDL 8_ DAQ WEPVL 1.000 I2f SVPLLTDLA F000 II 7 If TDLAQ WEPV F 0.02 0:] I 6 f LTDLA QWVE 4 .003 TABLEXX-V22-HLA-B3501 -9MERS-
PSCA
Each pepide is a parinofSQI NO: 8; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptidle is the start position plus eight.
STA T SBEQUENCE SCORE;' 1 fSPLLTHLif'iOQO 8 f LAQWEPVL f100 S9 LAQWEP'/Lv- 0.C9600' 3 T IfVPLTLA-__f .200;, f77fPLLTHLAQW 6 .50 F 7: THLAQWEPV f02 TH LAWdE- T 00106- ,TABLEXX-V24-HLA-B3501-9MERS-
PSCA
Each peptide is a portion of SEQ ID1 NO: 8; each start position is specified~ the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
SFTR SO SUSEQUENCE SCOREJ I IALMYCT' 0.501 _LTMYVCTPV F001 7 VTVHD 0OQ WO 2005/014780 WO 205/04780PCT/1§S2004/017231 ITABLEXX-V24-HLA-B3501-9MERS-
PSCA.
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amnine acids, and the end position for each peptidle is the start position plus eight.
STR IUBSEQUENCE] SCORE E7T-jCTPVPHPDi P 11D0-..1 __MYVCTPVPH 1 0.001 TALXX-V25-HLA-B3501 -9MERS-
PSCA
Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight._ STR SU BSEQU ENCE. SCORE .f j i TTRSS 18.000 ,f 9 fSSIDTDPPA 11.000 iF j: L ,.T.TRJlS 10.200 -i sSIDTDP 0.07 5 Ifs fTRQISSIDT F0.010 4 f PTRQS 0.00 tTABLEXX-V25&26-HLA-B3501-1 iEach peptide is a portion of SEQ1 ID NO: 8; each start position is specified, the length of peptide is 91 amino acids, and the end position for each peptide is the start ___Position plus eight.
~[STAR. lSUB SEQUGENCE! FsCOqR IID 71 PTRQISSSD 0.00 3! 1TABLEXX-V26-HLA-B3501-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified,I the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
S TAR T [SUBSEQUENCE I SCORE; f PTQGS [2.000 GSSDTDPPA L1000 a 4SSDTDPPAD 6. M.23- I fRQIGSST 0.020 IGSSDTDPP 0.015 1 5 j QIGSSDTDP 11 0.010 3F I f TRQIGSSD 0,.03 i[TABLEXX-V27-HLA-B35o1-9MERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids and the end position for each peptidleI is the start position plus eight.
START I SUBSEQUENCE: SCORE 2 ALRRA 0.010 A I LE X X I V 1- HLA--B3 5 01 10M E R S-
PSCA
Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, end the end position for each peptide is the start position plus nine.
START SBSEQUENCIE, CR lTABLEX(XI-V1 LAB30-1 OMERS-1 Each peptidle is a portion of SEQ ID NO: 2; each start position is specified,I the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
S-TA-RT IfSBEUENCEI [SCRE: L 1-1j. 271 AQ-vSNED-CL 6.000 69 NCVDSQDYY 4.000 98 iLA-QAAAL 0 F62 T .SGSLC Ij Ao ]fs IALPA 3.000i F]o107 'f LALLPALGLL Mf3000 13 YfLLPGTALL IF3.0001 F68-1 JLNGVDDSQDY IL .609 K__77-CC-D L [2.ooo FjI 1_ QPGTALLCYS [2.0001- 15f 7 QPTALLCY 52 4RIRAGLLT .[1.2-00 1I0. f.LLPAAA]LAL [1.0001 114 [GLLWGFGQL 1.000 106_ [ILALPALGl- j ~o 1- 4 VLA-LG D[1.000 1 I 9 f TARIRAVGL 1i.000 I12 IfGLALQPGTAL [1.00 54 fRAVGLLTVIS 0 0600 2. KAVLWLALLMVA. 0.60 1109 ,LALGLLLVV _j0.500 ASGAHALQPA[ .0 ,'-407 NCTQLGEQCWjfd765 7 119 11 TALLCYSOKA T0ol00 9 F AALPAA] .300 103 ~fAAAILALLPA [00 81 5 .l TCCDTDLCNA 0.3S009 29 QVSNEDCLQV f0.300 TFABLEXX-V25&26-HLA-B3501- 9MERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is9 amino acids, and the end position for each peptide is the start ____position plus eight.
START; ISUBSEQUENCE. SCORE F77[ PTQIS 'p F2.000 IfSSSDTDPP F6 .075 IF7 RQISSSDTD[00 1. QISSSDTDPI 0. '010 3 ,,TRQISSSDT Foc6 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 T ABLEXXI-.V1-HLA-B3501-1
OMERS-
Each peptidle is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each pepid isth strtpostin lus nine.
STR SUBSEQUENCE SCORE FL2i I[ LLCYSCKQ [90 0200 QCWTAIRA 0.200 ,f110 [LPLGLLL4WG [0,200 If4 417 1. CT QLEQCWT [0.150 L 3[ NiTCDTDLC. [0.150 47[ LGEQcwTARK[ F1i2L CVDDSQDYYV[ 0120 [ALQPGTALLCI[0.100[ f 58 LLTVISKGCS 10100 73 DSQDYYVGKK [0.100 I F LLALLMAGLA 0 If- 114 TQLGEQCWTA[ 1 0.10 84 [ITCCDTDLCN 0.100 IVISKGCSLNC 0100 F6i7i G -CSLNCVDDS coo If22-1 0C.CAQS 00IO 1G ALqP AA 0.100 7 f YVGKKNITCC 00 I DLCNSGAHA 1001 EQCTAR RALoc VSNIEDCLQVE A0.100 99, iF ALPAAAILA 6f00 F-57 GLLTVISKGC .1 00i TVISKGCSLN [0.1900 I3 fENCTQLGEQCO.0 11 AGLALQPGTA 00 31 FSECQEN 0.060 24 YSCKAQVSNE 0.050 1E7~ 4'_iCSLNCVDDSQ 1 0.050 9f_AHALOPAAMI f0.040 4QDYYVGKKr'U 0.040 53 4IRAVGLLTVI f04 42 [SCKAQVSNED 6f 030 071NASGAHALQ 4 03 [TABLEM4-1-HLA-133501-10MERS- Each peptide is a portion of SEQ ID NO: 2; each start position is specified, I the length of peptide is 10 amino acids, and the end position for each peptide is the start posiflon plus nine.
ST7ART[SUBSEQUENCE SCOREI 86 JKP DTDLNAS T 0.03% f88 [TLNAA 0 ~L~LLQVENCTQLG 419?9] F. 7.I KGCSLNCVDD F 37 LEQCWTAR F[ 0.020I 0.015 67 fSLNCVDDSQD 1 0.015.
[:1bo5 If AILALLPALG [o0.0o _RY.I YYVGKKNIf 0[.010~d 13_ I DCLQOVENCT[0' O10 f26 f1 CKAQVSNEC7,0.0!06 If5 fVCLLTVISKG 10~oi V112 4AILGILLGk 001 711 AVLLALLMAG 0.1 E-5- 7AVGLLTVISK 0.010 9..2.II LMIAGLALQPG 0o.010o 7i ARRAVGLLT I .1 20 ;fALLCYSCKAQ 4 .i0 '7 7[ CNASGAHALQ 4 .010 8Ff73 LLGLL Q 0 4.010 F 7 ALLMAGLALQ F0-1 YYVGK'KNITCf .i 23 CYSCKAQVSN fo 0oi TABLEXXI-V4-HLA-B3501-1OMERS- -P-SCA Each peptide is a porticn of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
F§TARTIl SUBSEQUENCE ,SCORE' TABLEXX-V4-HLA-B33501-1 OMERS- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptidle is the start position plus nine.
ISTAT SUBSEQUENCESCR I100 [_APVPHPDPPM 40.00 176 SPSRQL 27 MCSLPS 20.000 10 VHPDPPM AL ]0.O- 174-J CS"ASR 1000 _J 13 f __7VPEAHPNASLi600 8 7 EAHPNASLTM _IF.00 506 4AYLWGAPL If5.000 J F[1 PAF'STLNPV 4000 F7'FPLQPGVV 400 1 so4HPNASLTMYV[~00 2[27- :KPPADGsPiT-I 40001 F43 iACCSGIDPASY [_3000 1 7~[QPTLGVVQ .00 4 66 VPQAVPQA-i2.00 311RLPPISL:RCSL II2.000 F48lDPASYRLWGA 2.000 I1 TSRAVTPTCA_410 [[29 CSRLPIPSLRC .1 isco 117-1 4 CCFPF 100 1854 VVPQASVPLL I 1 .000 TLNPVLRH L [.00 [64 [1l GVVPQASVPL I7o 145 4STLNPVLRHL 1000 '7 11 WARRTSIRAVT71[ 6oi 1113 RTTRQGSl...0.809 56 7GALQ5PTLGVI 0.600 I..AVPLLHPA oo [75I SPAPSRGQ A~ b 4 41 H HS ACCS §GDP-A 0F 0 7155 IQEF-PAHTJ-IP7 .9, WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TABLEXXI-V4-HLA-B3501-1OMERS-1
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each Ipeptide is the start position plus nine.
{START!
L
T L-SRTPTRQ' I 0400 L LRCSLHSAC 0.300 f207 CA.GPP 300 91 ~.'NASLTMYVCA 0 300 L142_ PAFSTLNPV 0.300 110 VLRHLFPQEA j 030 42 SACC-SGD~PAS 0.300 23 PGPPCSRL 0.300 173 LWG PLQPT 0.200O 32LPPISLRCSLH 1110.200 JF74 L[THPA-WEPV F 0.2007 148 NPVLRHLFPQ 10.200 81 F VVPEAHP 0.200 84 L EAHPNAS4 .200 16 DPPMALSRTP 0.200 GPMPCSRL-PP 0.0 33 PPSLRCSLHS .0 178 A"PSRGQALRR7 0.200 22IF -MPS 0.200 4 RTTTWARRTS [00 F 1 7F TPTCATPAGPOI' 0 200 {93 [SLTYVCAPVf020 37 fRCSLHSACCS 0.200 133 ['_FLCCCFHGP 00 :f 62F[TLGVVPQASV. FT 0200 {157f QEAFPAHPIY [0200 78.AHPNASLTMY 0.200 .PPMALRTP 0.200 14fPTRQ1GSD [0200ob~ 120 f GSDTDPD 015 46 IfSGDPASYRLW 0150I 1Il LSRTPTRQl 1 150 13-1 HLFPQEAFPA 05 [t TABLE)(XI-V4-HLA-B3501 -1 OMERS-
I.....PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino [acids, and the end position for each jpeptideis the start poitious rnpe, ISTARTIj S UEN CE ]fS OEi 17A9 1 1 PSRGQALRRA' 0.150 15 AVTPTCATPA_[010 11_IGSIDTDPPA 0.100 DGPSNLCCC 0.100 21, [ATPAGPMPCS IFTo6ioo 1~lLSQPyw8YsI0.100 I- 138 IF- FHGPAFSTL If 0E.5:0 11I9F AFPAHPIYDL 0o.1006 f SQ'/WSVVSPA if too ~TiIAQwEPvL F6.667 136ji CCCFHGPAFS oo 137 iFCCFHGPAST~ F0. 100 104 H-PDPPMALSR 0.06 1" 4j RAVTPTCATP I .060~ 38 TCSLHSACCSG_ 0.050 172 7 WSVVSPAPSR 06.050, i 92 U gSYCAF 0.0506 1447 FTPRH [0.050= -O7[LsqvVswVsP 0. 050 3477 PSLRCSLHSA [0.050 158 [E f PAHPIYD. 003 62f AHIYDSQV 0.030.1 ~K~~fCAVPHPPP__0.030 T 68 f QASVPLLTHP 0.030 TABLEXXI-V19-HLA-B3501- 10MERS-PSCIA Each p eptide is a portion of SEQ ID NO: 8; each start pcsition is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
1STARTIi _SBSEQUENCEISCOREf 4 [MPCSIRLLPSL j ,20.000f1 8 fRLLPSLRCSL I2.000 10 ;LPSLRCLH i 2.000 CSRLLPSLRC ]f i ___AGPMPCSRLL if 1.000 4 PPC LLP of00 9 fPMSRLLP 0.010 45 CSRLLSLR .010 ITBLXX-v0- LA- 010ES
PSCA
Each peptide is a portion of SEQ NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each paptide is the start position plus nine.
lf STAR T SUBSEQUENcE IFfqCOR E [77 CSGDPASSRL 7T Fs -LwGAPL 6 fJ DPASSRLWGA 2.000 79 EfGsEE Q F -0.150! i 1ACCSGDPASS 150 IT4 GDASSRLW~ [0.101 i TABLEXXI-V21 -HLA-B3501-1 OMERS-,!
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, jthe length of peptide is 10 amino acids, and the end position for each ~peptide is the start position plus nine.
!START [SUBSEQUENCE SCRE! 10 f DPAQW 15j.000 f 9 TDPAQWEPVL Th0[o IF- 8 LTDPAQWEPV 0.060 i 2ifqASVPLLTD .3 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 [TA LEXXI-V21-HLA-B35a1-1
OMERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino I acids, and the end position for each peptide is the start position plus nine.
STAT ISBSEQ UENCE SCORE'- E1 7 4_LLTD PQWP {0.04 i 4 VLTDPAQ 1 0.010 6 4PLLTDPAQWE 0.001 TABLEXXI-V21 &22-H-LA-B3501- 1OERS-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end positicn for each peptide is the start position plus nine.
[START SUBSEQUENCESCR F41VPLLTDLAQW 1115-000 1 ASVFLLTDL 13000TO.
124 A VLL 405300 I _TDLAQWEPVLJ0.100 F 5_4 PLLTDLAWE[0.001i ,ITABLEXXI-V2-LA-B3501-1 OMERS-i
PSCA
Each peptide is a portion of SEQ 1ID" NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each 1eptide is the start position plus nine.
I START, rf UBSEQUEN67f!SCOREI I- T7 F- VPLLTHLAQW 10.066 I~ 14 -A-SVPLLTHL ii 300 KF 2 APLHLA F 050 I HAQWE PV.LV 0o.300i 7 L-THlAQ-WEPV 200b 8 4 THLAQWEPVL 7E.100 QEPVLVF P .3 I 31 4 .SVPLLTHLAQ 9.1 TALEXI HLA-B3501-10MERS-1 Each peptide is a portion of SEQ ID7 NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.] [START SUBSEQUEN CE SgCORE [61LLTHLAQWEP 0.010 5 P--7LLLTHLA QWE _0.001 [TABLEXXI-V24-HLA-B3501-IOMERS-1
___PSCA
Each peptide is a portion of'SEQ ID NO: 8; each start: position is specified,, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
STRTSUBSEQUENCE' SCORE _10 TPPPDPPM 4.0 1L NASLTMYVCT 01[.30 4 3 SLTMYVCTPv j 92067 2 ASLTMYVCTP F6.050 9 TMVCPDP 10.010 [CTPVPHPD7PP 0.9 Th1 TM ~PP4.010] I[ LK V TVPHP 0.1 [TABLEXX-2-LA-B3501 -i0MER A Each peptide is a portion of SEQ [D NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus ie STAR SUBEQUENCE SCR -LsRTPTRQIS j 1.609 3 Ij RTPTfRQIS fi-oo 0- 9 ISSDTDPPA4 .6 4 TPTRQISSID ~f0.20 4 7 __RISSDTD__40.020 1, 8-[QISSIDTDPP_ 0.015 2 [7 7 SRTPTRQ]S.010
I
184 TABLEXXI-V25-HL11A-B3501 -1 OM ERS-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptida is 10 amrino acids, and the end position for each peptide is the start position plsnie F-START [SUEQUECE ISORE 4 8 TRQISSITD 0.001 TABLEXXI-V25&26-H-LA-B3501iEach peptide is a portion of SEQ ID NO: 8; each start position is specified,I the length of peptide is 10 amino acids, and the end position for eachI peptide is the start position plus nine.] 'fSATIf SUBSEQUENCE.1~ SCE 7__LiJSIPPA 0 .500 'I 1[ RT-PTRQISSS_ 1 1J.200,_ f_ ][TPTRQIS SSD S8 4SSSDTDPPAD ;[715~ I 3 FPTRQlSSSDT 40.030~ L6 QISSSDTDPP 4.1 TRQSSSDTD Foo ITABLEXXI-V26-HL11A-B3501-1
OMERS-]
1 PSCA- Each peptide is a portion of SEQ ID 7 INO: 8; each start position is specified,1 the'length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
SAT SBSEQUENCE S-COREl I IRTPTRQ-IGSS 2001 2 TPTRQIGSSD 40.200 GSSDTDPPAD Pf..150 7 1fSDTDPPA I I 31 -TRQCIlGS SDT 0o.030 5U ~RQIGSSDTDP__ 0-.020 1 ,,,,,SSDTDPPADG 001 6 QIGSSDTDPP !F0.01 71FSDTDPPADGP1 0,002 TALEXXI-27-HLA-B3501-1 OMERS-.j WO 2005/014780 PCT/1§S2004/017231 PSCA acids, and the end position for each Each peptide is a portion of SEQ ID l .peptide is the start position plus n -ine.
NO: 8; each start position is specified, I TR SBEUNC.CR the length of peptide is 10 amino I ~E PS RGQ LRR 015 2 SRGQALRRAQ 0.001 if WO 2005/014780 Tables XXII XLIX: PCT/US2004/017231 TableXXII-V4-HLA-A1- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 Isore 123iDTDPPADGPI 21 15811 EAFPAHPIY 19 F04 HPDPPMALS 17 44 CSGDPASY 16 -4 SGDPASYRL 16 79 QWEPVLVPE 16 891HPNASLTMYI 16 1451 STLNPVLRH [16 I0 SRLPPSLRC 13 179 PSRGQALRRI 13 12T1 SDTDPPAD 12 127 1PADGPSNPLI 12 VPEAHPNAS 11 S34 PSLRCSLHSI 10 61 PTLGVVQAI 10 TableXXII-V4-HLA-A1gmers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
PosI 123456789 sore 67 PQASVPLLT i 1 74 LTHPAQWEPII 1 ~78 AQWEPVLVPI 10 105 PDPPMALSRI 113 RIPTRQIGS 10 156PQEAFPAHPI 1 165 IYDLSQVWS 10 TableXXII-V19-HLA-A1- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 |e 2]PMPCSRLLP| 91 6 |SRLLPSLRC 9 L llGPMPCSRLLI IMPCSRLLPSI 6 l|CSRLLPSLR I 7 RLLPSLRCS I TableXXII-V20-HLA-A1- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Po] 123456789 score TableXXII-V20-H LA-A 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 3 SGDPASSRL 16F 5 DPASSRLWG 7 8 SSRLWGAPL 7 TableXXII-V21-HLA-A1- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 s 7LIDPAQWEP I ASVPLLTDP 9 TabeXXII-V21&22-HLA- A1-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 score I 61LIDLAQWEPt 161 I11|ASVPLLTDL 9 I3 VPLLTDLAQ1 7 TableXXII-V22-HLA-A1- 9mers-PSCA WO 2005/014780 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end poSi on for each peptide is the start position plus eight.
Posl 123456789 Ej 1 ASVPLLTHLI FiI F3 V PLLT HLA() LI7 [Table~XXI-V24-HLA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
Posl 123456789 k F-3 LIM CT 8 VCT7]PHP7 Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
FPos 13578 cr F-2] RQS Ii~~ F-1 E]PT~I F-4] TEP E~ Fj TQISIT I ~IDQPLj TableXXII-V25&26-HLA- A1-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posj 123456789 ILSSSD-TDPP]114] [TableXXIIV26-HLA..A1..
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end P osition for each peptide is the start position plus eight.
[rableXXIIV27-HLAA.- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end Position fr each peptide is the start position plus eight.
[P-osI 124578 R F11 FEGA]2A TableXXI II-VI -HLA- AJ2O1 -9mers-PSCA PCT/1§S2004/017231 Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 14 ALPTAL 108ALLPALGLL 105 ILALPAL 29 F-ALLAGLA 26 Fl115 LLWPGL 26 109 LPALLLL 24 53IAVMT 23 F7 LLALAQ 21 20 ALCSCA 21 1'07 [=LALL 21 F13 FAQGTL Fi MKVLL 19 VLLAL4A 19 [12 GLL T 19 F54 RVLTV 19 57 GLTVK 19 TSKCS 19 10O2 PAAIL 19 4 3 QLEC 18 51 ARRGL 18 98 AQ 1AA 18 101 QPALA 18 11 2 FLLLW 18 AVLLM 17 50~ TAIAG 7 F63 SKCSN 1 7 83 NITCCDTDL 1 7 104 AALLP 17 106 I L 17 F22 LCSKQ 30 VSELQ I: 67 SLNC-DS WO 2005/014780 TableXXIlI-VI-HLA- [A0201 -Smers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight. j [Pd 123456789 I1r 114GLLLTVGPGQI 15 F361 LQENIT 14 r48 CWTA A 145 F46SGDPASYRL 15 127 PAD GPSN PLF 15 139 FHGPFSTL15 153 HLPE F 151 1163 FPYDSQ 1 5si F39 FLHA1S4]7 F51 SYL'GA 14 53 LGPLP 14 F61 PTGVPA14 SVLTP 14 F 83 VLPAP 14 8 6 PEHNS 14 143 AFT PL 14 145 STNVR 14] 24 AGMR 173 76 HPAWE 3] 77 PAWPVV 13 84] LVEHN] 13] 114 TPRIS 13 150 V HLQE 13 J 1 0 EVSVSP 91 ITableXXII-V4-HLA- [A0201-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
PosI23468 soe TabeXlllV4HLA- A00 -Pmr-SCA Each pepilde is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end psition for each peptide is the start position plus eight.
Pos 123456789 146 TLNPVRHL 2 F57APLQPTLGV 1 160 FPH1D 19 167 DLQWSV 19 F55 WPLPT 18 [94 LTY1P 18 F63 LGPQ 17 6542 FVQSP 17 [31 FLMVA 17 3166 FLS1C 1 668 VPAVL 16] 32 LPSRCL 15 F46 SGPS 127 PFGPNP 13 9 HP FST 153HFQF 163 HPY Q [39 SLSCS 14 F51] SYL P 14 [53 RLG Q 14 611 PFGVPQ 4I 7 0 FVLP 14 [83 FLVPAHP 86[PEHPNSL 4]j 143 AF14PV]I~ 145 STFPLR 4 24V 13]CSLIih F76 HPAW3PL 77~~F 3AWPVVj 84LPEAHPA 188 PCT/1§S2004/017231 TableXXIII-V4-HLA- A0201 -9mers-PSCA Each peptide Is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[PosIl 12345678 1 11 [LSRTPTRQ 1 ,14 TPTRQIGSI 11 13 [1750 QVS P 13 TableXXlIl-V1 9-HLA- A0201-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each paptidle is the start position plus eight.
IFos] 124689[ 8~ 1-4
ENS~SLL~
RLPSRC 71 WO 2005/014780 TableXXlII-V21 -HLA- A0201 -9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positon fr each peptide is the start position plus eight.
Posl 123456789 1 [1 ISVPLLTDPA 1 [7V6AQWE 13 F-1AQWPV 12 [7 PLLT AQ 11 F-7 FTP~E LiP [TableXXIIl.V21 &2HL-A-i A0201-9mers-PSCA] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[P os 13568 F Re [78 DLQ FPLJ 22 [21 ASPLrD 191 171 LLTDAQW [-2SPLTL 13 TableXXll l-V22-HLA- A0201 -9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 L HLAQWEPVL L PCT/1§S2004/017231 TableX)(lll-V22-HLA-1 A0201 -9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
P os 1 2345 6789- [211 ASVPLLTHL F19 Fj2] SVLTHA 14 F-5 FLHAQE 12 TableXXlll-V24-HLA- A0201-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[7 STMVT[7 [7VC [VPH 12 TableXXIII-V25-HLA-1 A0201 -9mars-PSCAj Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
ISI~j3 [A0201 -9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start -position plus eight.
7 2 QISIT [28] TableXXIII-V26-HLA- A0201-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start posiion plus eight.
Pos12468 F- [ElGSDO =7 [=SST] P 76 [4 =RI~ST[6 =1 RQIS DD][ WO 2005/014780 FTableXXllll-V26-HLA- A0201 -9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
F9 SDTDPPADGI[ -6 IGSSDTDPP L DPTRQIGSSD fj:N TableXXIII-V27-HLA- A0201-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
FTableXXV-V1 -HLA-] IA0203-9mers-PSCA A0203-9mers-PSCA [Pos 13579soe] NoResultsFound. j: TableXXIf-VI -H LA- A0203-9mers-PSGA [P-osI 24689soe ENoResultsFound.
A0203-gmers-PSCA Pos 13578 cr INoResultsFound.
TableXXIV-V21-HLA- A0203-9nlers-PSCA IPosi 2468 NoResultFound.
Table)0(IV-V21 &22- HLA-AD203-gmers-
PSCA
NoResultsFo-und.7 TableXXIV-V22-HLA- A0203-9mers-PSCA Posu! 4679sore TaleMVV24-LA-] NoResultsFound.
FTableXXIV-V25-H LA-] [A 0203-9mers-PSCA [NoResutond TableXXV-V25&26- HLA-A0203-9niers-
PSCA
PoE 135689soe [NoResultsFound.
ITableXXl V-V26-H LA- LA0203-9mers-PSOA FNoResultsFound.__ TabIeXXV-VI -HLA-A3- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
521Oj RIAVLL RLD 190 PCT/1§S2004/017231 TableXXV-V1 -HLA-A3- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Po s 13568 FVSK1S 1 115 6L] GQ 19 12 GLLG A 1 1065 IL LA 1 109 LLAGLL 1 438 FL-C~A 17 902dLNSA 17 57 [LTISG 16 105 EIAL A 1 114 GLLGPQ 16 LLALMAG 20 ALCCA 7 DSDYG 78 FVKNIC 67 SLC DS 1 94] A AALQI14] [112 ALLLW 14 I[2 VNE EQ A I 61]] 1KCSNI1 F-MA9 AQP1 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 FTabIeXXV-V1 -HLA-A3- L. 9mers-PSOA Each peptidle is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
7Pos 12 3456789 j 53IAGLLV 1 74 SQDY Y VG KK 1 16QRGTALLCY 11 F21][ LLYSCKAQ 11 54 RAGLTV 1 107 AA A=ILALL P 1 TableXXV-V4-HLA-A3- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
IFosI 1235689 f~~ 149 178 AP SR GALR 1 17 PRGQA LRR 1 F-9 ARRTSRAVT 1 44 CCSDPASY 14 65 WPASVP 14 73 L LT H P AQWE F14 146 TLPVRH 1 39q SLSCCG 13 F62 TLGVWE QS 1-3 6A SVPLTH 13 1 7 GSDD 1 32 DLTII A 109 MASRPT 2]~ 51SYLGAL I F881PALMf~ 118
FIGSDTD
121 IDTDP1AD abEXXV-V1 9-HLA A3 9mers-PSTCA I TableXXV-V21 -HLA-A3- 9 mers-PSCA FEach pptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus -eight.PF Pos! 123456789 scoJe WO 2005/014780 ETableXXV-V21 &22-H LA- A3-9mers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positon or each peptide is the start position plus eight.
Posl 123456789 Rj jjj4j PL-LTDLAQw] 18 5]LLTDLQWE15 I~L Q W E 152 Tab~eXXV-V22-HLA-A3- 9mers-PSCA Each peptide is a porticn of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positon fr each peptide is the start position plus eight.
Pos 124578 k F-4 ELTLQ 18 Fj-8 LQVEVL 1 F-51 LL1-4W SV2TL Tab~eXXV-V24-HLA-A3- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide s the start position plus eight.
PosJ 123456789 k j~ F-21 SLMVT 15 F-6]PPH 1 0 LIiJASTI'1V A MYGTVP 9 D~ TMVCPV 7 nTableXXV-V25-HLA-A3- 9mers-PSCA- Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, end the end position for each peptide is the start position plus eight.
Pos] 124578 F7 RQSIT 11 7 QISS I DTDP F11 F-2 RTTRIS 8 TableXXV-V25&26- HLA-A3-9mars-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[Pos12-3456789 se] LA TPRQSS i TableXXV-V26-HLA-A3- 9mers-PSOA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
124578 3sor RQ1SELTD PCT/1§S2004/017231 TableXXV-V26-HLA-A3-1 {9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[Po 123456789- s rl~ [i F0QGSSDTDMPID] FPRIS 6] TableXXV-V27-HLA-A3] 9mrers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[TableXXVi-V1-HLA-A26-] Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
1245 789 7 5-81 ARRVGL17 LI3] AVFLLA 16 36d FQECQ 16 16 QPGTALLCY 3ENCTLGQ WO 2005/014780 TableXXVi-VI -HLA-A26- 9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptidle is 9 amino acids, and the end position for each peptidle is the start position plus eight.
[Pos 124578 score [69 NCVDS 1Y76 AQVSNEDCL4] F33 EDLQEN 741 [lo0ll QPAAA 174] 1I02 PAAAILALL I1-41 F818ALAGL 1 F-MAVLAL 13 F59 LTIKC 13 8 NIT1CDT Ta b IeXXVi-V4- HLA-A26 -1 9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
Po-st 24679E 158 EAPHP 25 VVQSP 18 [123 DTP1G 17 173 FVSAPR 17 [64 GVPAS 16 170 QVSVSA 1 F81 EV P EAH 1 F89 HPALTY 1 [97 FVA H 1 145 STNVL 14 146 TL LRL 14 1I49 PLH F 14 AVPCAP 13 161 PTGVQ 13 I 7EAHP SLT 13 [TableXXVi-V4-HLA-A26- 9mners-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[Po-)sI123456789 scr 1106DPASR 3 F111 RTSRAVTPT[12 I 66 VPAVL 112l F70 SVL P 12 [84 LVEHPA 12 160 F HP 1D 12 TableXXVi-V1 9-HLA- A26-9mers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[Po0s 124578 R F-4 PCRLS 14 LLSL] S 11 F- ]GPMPCSRLL 97~ L M:PCSRLLPS 6 TableXXVi-V20-HLA- A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
PoIs 124689 R~ S3D]SSR 11, 5JDASRWGL1 :1 SSLWAP PCT/1§S2004/017231 TabeXXVi-V21-HLA-1 A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
Pos] 123456789 1 cr LI1PAWPLI1 Tabl eXXVi-V21 &22- H LA- A26-mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
D L Q E V 1 68 F 1svPLLTDLA 12 F DLAQWE 771 TableXXVi-V22-HLA-] A26-9mers-PSCA J Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
Pos] 1l23456789 I~e Dj SPLTL7h EA~SVLTHA 1 F-6 ITLQE F7 D~ HL Q W EP VL 7~ WO 2005/014780 TableXXVi-V24-HLA- A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 score 6 YVCTPVPHP 14 3LTMYVCTPVII 8 8 CTPVPHPDPIl 8 2 SLTMYVCTPI 7 A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 lscore 4 PTRQISSID II 2 1RTPTRQISSI 10 6 RQISSIDTD 10 3-]1TPTRQISSI I 8 7[QISSIDTDP] 7 9 SSIDTDPPA 6J TableXXVi-V25&26- HLA-A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 score 2]I PTRQISSSDII 2 TPTRQISSS|1 8 -4j|RQISSSDTD|1 6 PCT/US2004/017231 TableXXVi-V25&26- HLA-A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 lscore 5 QISSSDTDPI 5 TableXXVi-V26-HLA- A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 score I IPTRQIGSSDI 13 IITPTRQIGSSi 8 TableXXVi-V27-HLA- A26-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
osll 123456789 score li IISRGQALRRAII 5 TableXXVII-VI-HLA- B0702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 |score TableXXVII-V1-HLA- B0702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 score 101 QPAAAILAL 7 ALLMAGLALI 16 13 LALQPGTALII 16 14 ALQPGTALLF 105 AILALLPAL 107 LALLPALGLII 151 S50 TARIRAVGL 141 99 ALQPAAAIL 14 109 LLPALGLLL 14 16 IQPGTALLCYII 131 5 11ARIRAVGLL 131 52 RIRAVGLLTI 13 102 PAAAILALL 13 11081 ALLPALGLL 13 1101 LPALGLLLWI 13 1 MKAVLLALLI 12 SLLALLMAGL 12 28 1AQVSNEDCLI 12 S92CNASGAHAL 121 TableXXVII-V4-HLA- B0702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
posll 123456789 Iscore S57 APLQPTLGVI 24 1601 FPAHPIYDL I-24 S66l VPQASVPLL| 23 I 76HPAQWEPVLI 23 S321 LPPSLRCSL I21 WO 2005/014780 LTableXXVII-V4-HLA- B30702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[PosIl 123456789 Ijj 176 SPAPSRGQAl 19 10-2 VPPDM 1 F22TAPPS1 1 27 PAD GP SNPL17 104 FRPMAS1 106 DPMR 16 11-4 TETRiGSI 1-6 143 AFTLPV 1-6 163] HPYLQ 16] F24 G F 15 86 PEAPNAS 15 1 30 GPNLC 15 178 APRQL 15 48 DPSYLW 1-4 H D P A 14] 14 F STLP 1 jjj RRS9 VT1 F1 TfTATP A 1 F28PSLPL1 F55WALPL1 6 5 VPA L1 12 5DPDPN1 139 FGASL1 [14 8N HF 13] 155 PQE FPAH 3] FI11 RSATTKh F14 RTTCT 12 E9PSRP 1 F27 MPSLPS 1 F33] PLCL 12 F46SDAY 1 1 LWGPLQT 1 TableXXVII-V4-HLA- B0702-gmers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Po s 12345-678 9- F711 VPLLTHPAQ 12j 81 EPVLVPEAH 12 [85] VPAHNA 12 89HNSLM02 [1107] PPFLRT 12 177 LASG A 1 191 TCTPGP 1 [101-8 PVHPPP 1 F146 TLNVLR l]_ [j5 2 RL QE F 11 11[ _QAPAP 11 E ITableX<VII-V 9-H LA- L 0702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
FPos 124578 i~ ~J GPMCSRLL22 MPSLP3 PCRLS 13] PSR EAH1 EILLSLRCL 1 TableXXVII-V20-H LA- 130702-9mers-PSCA PCT/1§S2004/017231 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end pition for each peptide is the start position plus eight.
P os] 1I234-56789 kjj LIF SRLGAL 7 I ISGDPASSRLE 1-2 LTableXXVII-V21-HLA-1 B0702-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
[Pos 123456789~ ITableXXVI I-V21 &22- HLA-B0702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
[os 123456789e LI~1 DAQWEPV 13 TableXXVII-V22-HLA- 8 0702-9mers-PSCA
I
WO 2005/014780 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
I AS VPL L TH L F14 F7 VPLLTHLAQ 13 7~jHLQWEPV 131 D SVPLLTHL-A][: TableXXVIl-V24-HLA- IB0702-9rmers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positon fr each peptide is the start position plus eight.
[Pos 124689]oe 3L T mY v-CTPv Tabl11eXXVI -V2-5-HHLA-1 B0702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acd s, and the end position for each peptide is the start position plus eight EAJ SSIT811 TableXXVII-V25&26- HLA-B0702-9mers-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
-1 TPTRQis-S1 10o
F~ISSSDTDPP
60b eXXVI-V6HLA- 80702-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
I]TPTRQIGSS 10q FI 7 GSSDTDPPA 1 6 TIG SST D 7 TableXXVII-V27-HLA-1 B0702-9mers-PSCAj Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 aino acids, and the end position for each peptide is the start position plus eight.
FTabIeXXV-V-l-I-LA- PBC8-9mers-PSCAl PCT/1§S2004/017231 Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus -eight.
Pos 1234567897F~j 50FTARIRAVGL F29 F-6 TVISKG0CSL 101 QPAAALALI F18 F-7 ALMGA 17 F99] ALQPAAIL 16~ 108o8 ALLPALG-LL 1 10RN ALLL 1 11-5 EAWPGL1 13j LALQPGTAL 105 AILA-LLPAL L 107LLPLL 1 F83 NITCCDTDLI 14 102 ALL 14A STableXXVIII-V4-HLA- D 08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acdds, and the end position for each peptide is the start position plus eight.
177 P SGQL 23] 51l SYLGAL 191 F66 PASPL 18 160FPAHIYDL 18 3:2 LPPSLRCSL I6 14 6] FPLL1-6 3 3 P PS L RC SL H 1:48 N LHLP 2j MPSLP 14 35 SL LS 14 127 FGNL14 WO 2005/014780 TableXXVIll-V4-HLA- B08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
IPos] F123456789 1 F49PSRWA 1 11-4TPTRQIGSII 13 0 VRLPE 13 [46SGDPASYRL F12 WP VL 12 1 09 MSTT 12 1 11]LSRTPTRQI F12 8] RTS V[ ill F86 PEHNAL 1 [103] PHDPA] 111i [139 FHP FST][ i Table(XVIII-VI 9-H LA-1 B08-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight. I Pos] 1 23456789 E1~ 8] SL S F16 B08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
PoI1246898~ j:SR]W3P 19 FTahleXXVlll-V21_H LA- B08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
PosF 12345-678-9 DE A QWE FL 16 Dj VPLTPA F- TableXXVIII-V21 &22- HLA-B0-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[Po0s] 13579~ E[ rTableXXVIII-V22-HLAi B08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
PCT/1§S2004/017231 FAi ASV PLLT HL ETableXXVIII-V24-HLA- B08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and te end position for each peptide is the start position plus eight.
B08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 1245 7 TableXXVIII-V25&26- HLA-B308-9mers-FSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of poptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Po0s12468 jjJ 71 TFTQ7SS F-2] [TQIS ]6L~ WO 2005/014780 FTableXXVIII-V26-HLA B308-9mers-PSCA Each peptide Is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
P osI 2468 FLZ TPTRQIGSS F FTableXXVIII-V27-HLA-1 B08-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight, Tab~eXXIX-V1-HL- Bi 51 0-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
ll-os]l 123456789] R 13 AQPTA 13 F14 AQGAL13A F92 CFHA 313 F97 DHLP A [i oi1 QPE9LL[ 3 [FMKVL1L[ 2 LELA [12 36 LVENC Q 1 TableXXIX-VI-HLA- 0151 0-9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
7Pos 1123466789 99 FLPAAL 12 105 ILALPA 12 108 [LPLGL 12 115 FLW GL 12 j-7 ALMGA 11 2 8 AQSE 11 1 ARRVGL 1 F60TIKGS 1-1 102 FAAI 1L 1 1107] LALAG 11 I_83 NICDDI 1591 LLAGLL[ 10 [TableXXIX-V4-HLA- B151 U-0mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end psition for each peptide is the start position plus eight.
1103 PH PML 24 1I39FGAFTJ 23] 1152] HFQA 8 FGPQ L 15 F46 SG SYL 14 143 AFIN L 14 F24 FGMR 13] 1 21 PSRLPSLL2]1 PCT/1§S2004/017231 TableXXIX-V4-HLA- B1510-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
F65 WPAS 13 1601 PID L1i3 F66 VPVL 12 [86] PEAPN 1L27 127 PAG P 12 F32 L SRS 11 40q LHSAC7SG11 [75 THAWP 11 1171_PPSRQA 11 TableXXIX-V1 9-H LA- 0151 0-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Tab~eXXIX-V20-H LA- 0151 0-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptidle is the start position plus eight.
Pos512468 3] SG S r 14 I18 SSLWAP 1 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TableXXIX-V21 -HLA-1 8151 O-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positon for each peptide is the start position plus eight.
[Po s F12 345 67 8-9 [TablexXlX-V21 &22-H LA-] 8151 -9mers-PSCA] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[Po0s 124578 de [8 FLAQ1PVL ADSVPLTLFq~ Tab~eXXIX-V22-HLA-1 RI51 O-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789e F-8] AQEPV 15 ASVP1 TH 12 DTLEPV 11 STableXXIX-V24-H
LA-
Dl 151O-9mers-PSOA 1 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[PosI13579~e 74 AS7] VC EN~ STableXXIX-V25-HLA- B1510-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos12468 D RPRI E SdTP FI3 3] RQSS [I2 RISSDT =2I FTab~eXXIX-V25&26- HLA-151 O-9mers-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end postion for each peptide is the start position plus eight.
dosI 12345789 e FTQI S D 2i E TR ISS T Z 3JRQSST
LIFQISSTD
TableXXIX-V27-HLA-1 BI 51 C-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
P os 12468 R~i TableXXX-VI-HLA- B2705-9mers-SCA WO 2005/014780 Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 jk F51 FRRVGL24 54 RVLTI 18 F13 LALQPGTAL 1FI7 F53 FLLV1-61 105 ALLPL 16 10O7 LALLALG 1-6 F2]LLL 1-5 11 5 LLWGL15 19!9 TALYSK[14 F44 FGQWA] 141 Fai QPAA9LA F 14] 1701 ALLLL 1 F76DYGKI 1 F83NTCTL 1 F92 CNSGIA 13 F98 HLPAI 1 102 PA LL 13 F28 QSEDL 12 GECTR 112 TA AG 1 2! [57 GLTIK 172~ 1 TableXXX-V4-HLA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
F3 FRTWR 23 152 RHLF PQEAF 19 F30 ]SRLPPSLRCI1 155WALPL 1 178ASGAR 1 17 9PRQLR 1 1 46SDAYL 1 12 7PDPNP 145SLPLR] 16 0 FAPD] s 1I73 SVPPS] 5 181 RQLRR 1 F65VPAVL 1 F76HAWPL 1 105 PDMLS 1 11 6 TRID 14 [~JTWARTSR 13 [29 FSLPS 13] [45 CSDPASR 1-3! [661VPQAVP F 13 117 FQGDD 13 13 9 FHPF13 146 TLPLRL13 158 EAPHP13 F13 SRTC 12 23 FP S 12 322LPSLCS 1 PCT/1§S2004/017231 TableXXX-V4-HA B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is speified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
[Ps123456789 [36LRCSLHSO121 [44!C-CSGDPASY 1 5 -1 SYLWA L 1 103 FPML 12 151] LR DQ A 1 FI 0 HR- TWA 11 81 EVPEE 1 TableXXX-V1 9-H LA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Po 3568 EA~ 78 OPPSL 152 19 PCRLPLA 4 WO 2005/014780 B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 sr I~ ISRLWGAPLQI 16 I3SGDPASSRL- 15 EI2CSGDPASSRI 14 ~JISSRLWGAPL 12 TableXXX-V21-HLA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
os 123456789 score [IIDPAQWEPVL 141 SASVPLLTDP 1 7 TableXXX-V21 &22-HLA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos] 123456789 Isco I ASVPLLTDL 1-17 7I8DLAQWEPVLIf 14 TableXXX-V22-HLA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posll 123456789 s [I IASVPLLTHL I 17 HLAQWEPVLI 14 TableXXX-V24-HLA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos] 123456789 |c F5]MYVCTPVPH 10 ~1 ASLTMYVCT [1 TMYVCTPVP 4 B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos1123456789 core 5 TRQISSIDT I13 I 1 SRTPTRQISI 11 7 TPTRQISSI 11 I61RQISSIDTDI 11 7 RTPTRQISSlI 8 TableXXX-V25&26- HLA-B2705-9mers-
PSCA
PCT/US2004/017231 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 Iscore I| TRQISSSDT113 I411RQISSSDTDII 9 2|PTRQISSSDII 6 TableXXX-V26-HLA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
os 123456789 ore SITRQIGSSDT 14 I 4|RQIGSSDTDII 11 TableXXX-V27-HLA- B2705-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 ls 1 ISRGQALRRA[ TableXXXI-VI-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos] 123456789 lscore WO 2005/014780 TableXXXI-VI -HLA- B2709-9mers-PSCA Each peptidle is a pcrtion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position fcr each peptide is the start position plus eight.
Posj 123456789 JE F51]AIAGL 2 F53 IFVLLV 20 77 ALMGLL 14 105 AIALA 14 1l07 LALAG 14 [158 AB PAGL 14 [i2] KALALJ13 [28 A-SE~L 13 54] AGLTIJ 13~ F99! ALP I 13 L WPQ 13 F13 FGA 12 F14 FLPTL 12 F22 FCS Q 12 3 6 LQEQ 12 FECTAI 12 TAFRVG 1 TVSGCL 12 92 ONSAHL 12 VFEDLQ 11 76 FYYGKI 1i 8 3 NECRTL[ 101 FPAALA 11 102 PAA FL 11 TableXXXI-V4-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
PCT/1§S2004/017231 Fpos123456789 F10o RRSAT 18 [30 RLPPSLRC F15 F52YLGPQ1 152 RFQEF 14 46 SGPSYL13 [24 AG12S F57 AQFTG 72 [66 VFAV] 12 [111 RPTQI 12] 143ASLPL1 16 YILQVS 12 F28 FCSLPSL11 32 LPSLCll1 6 5]QEPL1 [139 FHPFll 142PFTNV 1 146TNVRL1 F163HIDSQ] i 11777PPRQA i F13SATTC o F51]SRWAF 0 F88APALM1 10O3 FHDPA 1169RISIT1 F19 TCTAP 9 777 AWPVV~ [101 PVHDPM 91 ill1 LSTTRI[ 91 L111NPLCCFLJ 202 [TableXXXI-V4-H LA- B2709-9niers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end positon for each peptide is the start position plus eight.
136CCFGA 1757 QEFPHP 147 LPLHFLi TableX)(XI-VI 9-H LA-1 B2709-9mors-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
P osl 124578 LB2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
F7os 124578 ore 9 ISRLWGAPLQ LA~ D4PSRL7h SSRLWGAPL WO 2005/014780 TableXXXI-V21-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
P-s 123456789 scr L |DPAQWEPVLII 11| I8 TDPAQWEPV TableXXXI-V21 &22-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 s S[ASVPLLTDL 13 []|DLAQWEPVL Lr11J TDLAQWEPV 1i TableXXXI-V22-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 sco [I ASVPLLTHL 1-- 81HLAQWEPVL r- LTHLAQWEPVII 1 |LAQWEPVLV|| 9 TableXXXI-V24-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
os 123456789 Ilscore I ILTMYVCTPVI 8! ASLTMYVCTII 4 4 TMYVCTPVPI- 3 TableXXXI-V25-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
os123456789 score ZISRTPTRQISl 12 S5TRQISSIDT| I3FTPTRQISSII 9 -61IRQISSIDTDI 6 TableXXXI-V25&26- HLA-B2709-9mers-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
os 123456789 score S]iTRQISSSDTII SlRQISSSDTDll 6 TableXXXI-V26-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus 203 PCT/US2004/017231 eight.
Posl 123456789 I 31TRQGSSDT I i-
RQIGSSDTD
I] GSSDTDPPA 3 TableXXXI-V27-HLA- B2709-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 s 1 ISRGQALRRAI 11I WO 2005/014780 TableXXXII-VI-H LA-1 B4402-9mners-PSOA Each peptide Is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
109 LLPALGLLL F13 115 LLLWGPGQL F1 3 [7MALLL 12 36 QEN1Q 1 689 NCDS 12 CVQY 12 F98 HLPA 12 G 11 [38VNT G 11 [41 CTLEC 11 F54 R LT I 1I 8-3 NTCDD 11 F76 FYV N 10 101Ai LALLPA 3 TableXXXII-V4-H LA- B4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position pius eight.
pos 1 123456789 F86PAPAS 0 1157 QEFAHI{ 201 1168 EAFHI] 171 143 AFSTLPVL 1 F46 SFPSYL[ 151 110 3 PHP PML] 151 1139 FP FST][ 151 1146] FLPVRH] 15 F241 FGCR] 141 WGPPT 14 72 P(LLT AQ 141 TableXXXII-V4-H LA-] Each peptide is a pcrtion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position pius eight.
FPos 124578 g 1l47 LNPV/LRH LF 14 177 FSR1A 14 28 PCRL 1S 13 F44 CGPAY 13 F47 GDAS L 13 127 PA SP 13 131 PNLC F 13 152 RLP E 9 1 160 FPHID 13 32 LPSRS 12 65 FVQSP 12 6 6 VPVL 12 F80o WEVVE 12 111 LSTTQ 12 114 TPRIS 12 136 CCHPF 12 164 FID QV 12 76HPQ E E 1 89 HPNALTM l1 TableXXXiI-VI 9-HLA-] LB4402-9mers-PSCA] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position pius eight.
[7 PCSLLPSL 13 LLPLRCSL 12 [7 RLPSLRC EII6 PCT/1§S2004/017231 B4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position pius eight.
EASRWGP F12 TabeXXXli-V21 -HLA- B4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position pius eight.
TableXXXII-V21 &22-1 LHLA-4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
P os 12345678 IF Tabie)(XI i-V22-HLA- LB4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each [start position is specified, WO 2005/014780 the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 1 234567897 R 11 FSPLH 17 1FLLH Q F14 ETableXXXI l-V24-H LA- B4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posi 124689]~ 17ASTYVT11 7F6 VIM4 CTVP F7 [TableXXXI l-V25-H LA-1 LB4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
FP-osI 124578 F-3] RQSS F12 F-6ISS T [17 jj1j STTQSF775 [A SSDTP E3 STableXXXII-V25&26- HLA-B4402-9mers-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
17 TPTRQ-IS-S-S Li D7QSST TableXXXII-V26-HLA- B4402-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight, FosI 2468 [7SSDDPPAE17 TableXXXII-V27-HLA- B4402-9mers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
FPosF 123456789] E7ALRA =4 F1 SGQLRA 11 PCT/1§S2004/017231 TableX)(XIIII-VI-HLA- 66101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 9 amino acids, and the end position fr each peptide is the start position plus eight.
F~sF 23456789-[1 RAFVGLLTVIIlE 26 F 9-1 HLPAI 3 107 E9LALL 2 E9 LAQ T L 2 E9ARRG 719 LZtl DYVKN 19 10 QPAIL 19 1 0 2 PAAAL 1 [7LLLAGL 1 11L GLLLW [FQPGTALLCY 1 [56 FALYSK 13 [D VGLVS 132 II~1MALALQP 12 II~VSEDCLQ 12 10-o3 AILLL 12 11041 FALLP 12 TableXXXIIII-V1 9-H LA- B51 01-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Po0s 12468 PPGRL 17 :7 MPSLLS 13 :1 E L DLRSL LI WO 2005/014780 TableXXXIIII-V19-HLA- B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posll 123456789 Ilscore 8 LLPSLRCSL I 9 B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 score S51DPASSRLWGI 16 3 SGDPASSRLI 13 I 6PASSRLWGA 11 TableXXXIIII-V21-HLA- B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 [sco F91DPAQWEPVL 23 1 QASVPLLTD| 14 4| VPLLTDPAQ 14 S81TDPAQWEPV 11 TableXXXIIII-V21&22- HLA-B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide PCT/US2004/017231 is the start position plus eight.
os 123456789 core il VPLLTDLAQ I-15 I DLAQWEPVLI 1 71 TDLAQWEPV I12 I 1 ASVPLLTDL I[ 8 TableXXXIIII-V22-HLA- B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posll 123456789 Iscore 9]ILAQWEPVLV 22 3i VPLLTHLAQ 15 71THLAQWEPV I12 TableXXXIIII-V24-HLA- B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
IPosI 123456789 score I 3LTMYVCTPV 713 I 41TMYVCTPVPI 8 I 1YVCTPVPHPI 6[ TableXXXIIII-V25-HLA- B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
S112345678911score ITPTRQISSI 22 206 TableXXXIIII-V25&26- HLA-B5101-9mers-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 Iscore 11TPTRQISSS|l 121 TableXXXIII-V26-HLA- B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Posl 123456789 Isc I 1ITPTRQIGSS 12 611 GSSDTDPPI TableXXXIIII-V27-HLA- B5101-9mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 9 amino acids, and the end position for each peptide is the start position plus eight.
Pos 123456789 scre I271RGQALRRAQI I IISRGQALRRAI TableXXXIV-V1-H LA-A1- WO 2005/014780 Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each pepies the start position plus nine.
PosI 1234567890 E LQPGTALLCY] -22 37Q1 NTQG 16 69. NCDS F 15 74 QDYYV G KKN 16 1081_ALAGL F 15 14 ALPT L] 14 -71 VFSDYV 12 84 ITCTD 12 F99 ALAAL 12 32 NECL1-NC 11 F44 LGQWAI 10 F51 ARAGLT 10 TalXXXI V-V4-HLA-AI-1 Tabe mers-PSCA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide isl10 amino acids, and the end position for each peptide is the start position plus nine.
123 DTPAGS 20 46 SGPSYL 18 43 ACSDPS 16 79 QWPLPA 15 F157 EFPHPY 15 F127 PAGPNPC 14 185 PEHP 1S 12 121 SIDtDAD 12 [RI4 FSTLNPVLRHLi2 TableXXXIV-V4-HLA-AI 1 Omers-PSOA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptude is 10 amino acids, and the end position for each peptide is the start position plus nine.
Tos 1l234567890 OPPSRP Ill 1j2GSELC 8] 165IDSMSI i 29 COSRL-PPSL-RO][ 112 SRTPTRQIGS][ 761 A11 PIQI-l1 101 [156PEAFHII 10 F11Ii PAHPIYDLSQ If 10 [66VQSEL E 9 TableXXXIV-V19-H L1A-Al-i 1lOmers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos124680 sre F72 MCRLP 3 D ~LPSR 0 F3 MCRLP D LFLCLZ~ 11 LRCSLS IA TableXXXl V-V20-H LA-Al-] 1 Omers-PSOA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
:Po0s 1235679 i~e] PCT/1§S2004/017231 TableXXXIV-V20-HLA-A1-] lomers-PSCA Each peplide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
101 SLGALQ L18 TableXX(XIV-V21-HLA-A -1 l0mers-PSCA J Each peptide is a portion of SEQ ID NO: 6; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
=Pos 1245780 =8 LIPA2E 0 3 ASVLLDPALI Tabl eXXXIlV-V21 &22-H LA-i Al-i Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
[pos~~ 1245780 6e [jASV-PLLTDLAQ]I81 [Ta bleXXXI V-V22- H LA-Al L 1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of poptide is amino acids, and the end position for each peptide is the start position plus nine.
F1234567890 scr WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TableXXXI V-V22-H LA-Al 1 Omers-PSCA- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine, 1 ASVPLLTHLA IF11 TableXXXIV-V24-H LA-Al 1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Fos 35 679 E TPVHPPP =4 D1 SLTYVTP =3 TableXXXIV-V25-HLA-1 Al-I Omers-PSCA Each peptde is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amrino acids, and the end position fr each peptide is the start position plus nine.
1 RTPRQSS TableXXXIV-V25&26- HLA-A1 -1 Omers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is amino acids, and the end position for each peptide is the start position plus nine.
F71 FIRISS 6]1 [TableXXXI-V26-HLA-Al-] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
Pos 124679 [A SDTDPAD 16 TableXXXIV-V27-HL11A-AIl- 1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
Po s 123458789 17 F-5QLRA1 17 RGALRA [13 TableXXVV-LA A0201 -11OesPC Each peptide is a portion of SEQ ID NO: 2; each start position is specified: the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
PoS! 24679 108[ ALPL L A WO 2005/014780 TableXXXV-VI-HLA-7 A0201-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each pepide is the start position plus nine.
Posl 1234567890 ]F ALLCYSCKAQI 14 1I05ALLPL 14 Kj]AV-LLALLMAG 173 F43 QLE FA 1j31 F57 FLT1SG FIB [67 SLCDDQ 1 TableXXXV-V4-H LA- A0201 -1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptidle is the start position plus nine.
POs 3 45689 F31] RLPLCL 24 F62 TLV QAV 24 F93 SLMVP 23 1 45 STNVL 22 F53 RLGA QT 21] FVQAV L 20] F 110 A RTTRI 20 F64 GVVQAVP 19 F57 FPQPhG 18 74 LTPAWEV 18 [83 VLPEH A 18 F113 FTRIGI 18 1 65 IYDS-S 18 F56 GAFPTG 17 141 GPFLNV 17 0 LHF 17 F153 H EFPA 1 [1569 AFAH L 17 176 SPPRGA EE7 TableXXXV-V4-HLA-1 A0201 -lomers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptidle is the start position plus nine, i124679 score] 12AHPlYDLSQV IF-161 76 HAQWEVLV 14 F85 VPAPNS 14 138 OFGAS l 1 146 TLPLHF 14 166 FDSQ S 14 FTW2 RT7]A 13 27 MCSRLPSL 13 3542 SFCLH 13 3:9 SLSACGD 1 45 9SDASR 12 79 QWPLPA 12 175 THPQWPV 11 78 AWEPVVPE 11 5 TYCPP 11 134 PLCF1 11 TableXXXV-V1 9-I-LA- A0201 -1 Omers-PSGA PCT/1§S2004/017231 Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is amino acids, and the end position for each peptide Is the start position plus nine.
F RLLP-SLRCSL F26 74 MPSLLS 17 D LLPSLRCS-LH 12j A02011-1 Omers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine, E ASSL P 14 LTableXXXV-V21-HLA- A0201 -1 Omers-FSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is amino acids, and the end position for each peptide is the start position plus nine.
L A QWE VL 113 TabIeXXXV-V21 &22-H
LA-
A0201 -1 Omers-PSCAf WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nne F1 QAVLLD 18 F77TLQEV 13 TableXXXV-V22-HLA- A0201-1 Omers-PSCAI Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each poptide is the start position plus nine.
PO s 124689 EcR F7 FLHAWP 161 7jJHLQW]V F13 LTableXXXV-V24-HLA- A0201 -1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
[PosI 1234567890 Ikje] A0201 -1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 34579 cr Tab~eXXXV-V25&26-HLA- A0201-l0mers-PSCA Each peptide is a portion of SEQ ID NO: 8; eaoh start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine, DR-TPTRQISS1 TableXXXV-V26-HLA-1 A0201 -l0mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine, Po-s 13579 [scorel DFR QISTDPj 81- D~lSDDPA[7 DE QIGSDTPI411 TableXXXV-V27-HLA-1 A0201 -1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is ICj amino acids, and the end position for eachppiei the start positionpuc ie D PSRGQALRRA 7IIZ FSRGQALRRAQ
L
L TabIeXXXVI-VI -HLA- A0203-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plusnie F96GHLBAA 2 90 LCNASGAHA 18 TableXXXVI-V4-HLA- A0203-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
PosI 124679 34L RCLS 41HSCSGP 48 DASYRWGA 83 V LVPEAHPNA 191 NALMYC 101 PVHDPM 119 E9IDPA 1 134 PCCCFGPA WO 2005/014780 TableXXXVI-V4-HLA-1 A0203-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length ol peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 124679 R 169 SQ VVP 10 175 SPPSGQ 10 17 9 PS LR 10 F13 SRVTTAT 9 F161 FTTCTA 9 F351 SLRCSLHSAC 1779 F42_S SDA 179 F61] FTGVPA 9EI 84
EVPAHNA
92 ASTMVA 97 102 VPPDPM 9 120 GSDD 9A i 135 LCCHPF 9 1 LH FPQAF[9 4 LQEFPA 9 170 FMSVSPP 9 1 76 FPPSGAL 180 RGQALRRAR EII 72 TLRITAR 8I~ F-8 RTSAV F778 F14 RATPCA 8 F17 TPCA8GP F36 =RLSCC1 [43 ACSGPA 8 5 ASRLGL 8I F62 TLVV SV1 F71] VPLT-8W1 F81 EPLVEH 81 [PAHNSL1 93 SLTYVP 17 Table(XXVI-V4-HLA- A0203-1 Omers-PSCA- Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide i the start position plus nine.
Pos F1234-5678907 F e 103 PHDPAL -18 121] S!TPPD 8Ll 1736 CFHPF 78 152 RHFQAF 18 155 FEEAFPAHP ZFi8 TableXXXVI-V1 9-I-LA- A0203-1 Omers-PSCA I NoResults~cound.
TableXXXVI-V20-H LA- A0203-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
[Po 12356890 re DPSRLG F77T [F PASSRL-WGAP F19 ~CSDPAS 18 [E SSLWAP 18 TableXXXVI-V21-HLA- A0203-1 Omners-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine, Pos 123457890 e =3 AFPLP 10 7 SVPLTDFQ [1 211 PCT/1§S2004/017231 TableXXXVI-V21 -HLA-1 A0203-1 Omers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end posiion for each peptide is the start position plus nine.
[jRVBLTDRQW 1 TableXXXVI-V21 &22-- HLA-A0203-1 Omers-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
7 ASVPLTDLA TableXXXVI-V22.-HLA-] A0203-1 Omers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus n ine.
Tab~eXXXVI-V24--HLA- A0203-1 Omers-PSCA Pos1234567890re N7oResultsFound.] A0203-1 Omers-PSCA WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position fr each peptide is the start position plus nine.
Posl 1234567890 IER~ 1LA9A0F-1-01 TableXXXVIlV25&26- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
TableXXXVI-V26--HLA- A0203-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
[PoS] 124679 R F7 F1STDP 10 TableXXXVI-V27--HLA- A0203-1 Omors-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
POS 124679 i j1[ E~GA~RA1 2 SEQARR q TableXXXVII-V1 -HLA-A3-1 I Omers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
[P-osI 1234567890 score F55AVGLLTVISK F26 F5-2 RIRaVG-LLTV 24 518 LPAGLL 24 F14A~GAL 22 F8 LLM-AGLALQP F-21] 106 FLLPA 201 F29QSE Q 119 F43 F1~WA 1 F9-91 F-AL 1 [105 AIALAGF 191 [3 AVLLLA] 18 [F ALM7] AQ] 181 1 14 GLLEQ] 18 VLALMGj 17 [12 GLQGL[17! [67 SICQQD[171 190 DLNAAH] 171 [751 LLLLAA] 161 [20 F1CYK 161 F21] LLYKQ] 161 [37 DNIQG] 1 TVF~L 161 [18GALXC] 151 F51] ARRVGL] isi [112AGLWP F35 C EN 1Q 14 [72 FDQDYV 14 F111 ALLPT 13 F57 GLTVSC 12 F73] F1D K 12 97 AHLPA 12 Tab~eXXXVI l-V4-HLA-A3- I Omers-PSCA
J
WO 2005/014780 TableXXXVII-V4-HLA-A3- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nne [PosI 12356790 e~ 143 AFSTLNPVLR F-12 F11 RLEATTC 1 F14 RATPCTP 1 F28 PSLPLR 11 TableXXXVII-V1 9-HLA- A3-1 Omers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
FPos1234567890 scr F-J LSL SL A1 [TableXXXV l-V20-HLA-A3- 1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
P os 12356790 e F72 SDPSS 14 F7 FSRWGP 12 SRE9PQP1 TableXXXVII-V21 -HLA- A3-1 Omers-PSCA__ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position pu ie Pos 1 234567890]Fj SSVPLLTDPAQ] 13 PQASVPLLT 0I1 TableXXXVI l-V21 &22-1 HLA-A3-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos~~ 1245780 6 F F1DAQE 11 TabeXXXVII-V22-HLA-A3-] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 10 amino acids, and the end position for each peptide is the start position plus nine., F-3 SVLTH 16 D7 PLTLAW 16 F-1 H QEPL 16 LI6] LAHAWE~ 1 DI VPLTLAW 9 F-1 THA FPL[7 10 LAWE=EPLLVP LZ PCT/1§S2004/017231 LTableXXXVI-V24--HLA- A0203-1 Omers-PSCAJ Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is1 the start position plus nine.1 Po 12356790 e [1JT VOP H F13 FA MVCP 12 F7]~VTVHD 1
ASLTMYVCT
ETableXXXVI-V25--HLA- A0203-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amn acids, and the end position for each peptide is the start position plus n ine.
Pos 124679 [D Q ESATPP1
[ENRTTQIS
LIF RIS7]P 7 SSDT7ADEZZ [:Al TPTRQISSID F7 TableXXXVI-V25&26--1 [HLA-A0 203-1 Om ers- PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.1 [Po-s 124680 i ~e DjISDDP EA~o [7gSSDDPADZ ITableXXXVII-V26-HLA-A3- 1 0mners-PSCA WO 2005/014780 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
F~s~ 1234567890 72 TPTRQIGSSD1 11 RQGSDD 101 F6 QISIJDP 10 71 RTTR1GS 7 TableXXXVI l-V27-HLA-A3-] I Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
PosI 12356790 e TableXXXVIII-V1 -HLA-1 A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
1I23456789 Iscre F59 LTI OL 22 TFSGCL 17 F88 DTLCAG 1 104 AAFLA 17 LQG LLY 15 TableXXXVIII-VI-HLA- A26-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 aino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 F 73 DSDYVK 15 78 YVKNIC 14 [100 6 LQPAAAILAL F14 107 FALAL 13 29 QVNDCQ 12 F39 ENT GQ 12 F82 KNTCD 12 101 QPAIL 12 LAMGA L F11 37 QVENCTQLGE 11 146 EQWAR 11 70 CVDQD 11 ALPAGLL 1[i KALALA[10 [12 G LQGAL[ 33 EDLVEC] 10 529 FIVLL 1] 10 68 NVDSD Tab~eXXXVIII-V4-HLA- A26-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peplide is the start position plus nine.
I~osI 13456789 64 VPQAVPL 23 VVQA PL 22 14E N PVLRL 21 123DDPDP 181 173 SVVSPAPSRG 1 17 16] PCT/1§S2004/017231 TableXXVI-4HA A26-1oesPC Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is ami.no acids, and the end position for each peptide is the start position plus nine.
88 AHNSLM 56789-- 8 1E P V P A H 1 4 87 EHPNALTM 13 129 DGSPC 13 [142] PASLNV 13 15ATTA 121 F43 ACSDPS] 121 [1-5-9APHID 121 [1-70 FVSVPP 121 [149PLHFQ I [174 VSPAPSGQ ii TableXXXVII I-VII 9-HLA-1 A26-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is am ino acids, and the end position for each peptide is the start position plus nine.
[P-os 124680 E] MPSLPS 6 4 TableXXXVII l-V2C-H LA- A26-1 Omers-PSCA WO 2005/014780 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 scre 6 DPASSRLWGA 13 D CSGDPASSRL 10 8|ASSRLWGAPL TableXXXVIII-V21-HLA- A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 score 7SVPLLTDPAQ 12 8 LTDPAQWEPV 9 9 TDPAQWEPVL DPAQWEPVLV 8 S1PQASVPLLTD 3 ASVPLLTDPA 5 TableXXXVIII-V21&22- HLA-A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 score 3 SVPLLTDLAQI 13 F1]QASVPLLTDL I12 8 TDLAQWEPVL 9 [7LTDLAQWEPVf7 8 9 DLAQWEPVLV| 8 TableXXXVIII-V22-HLA- A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 Iscore 3 SVPLLTHLAQI 13 ]1 QASVPLLTHL 12 8 THLAQWEPVL|| 9 -7 LTHLAQWEPV|| 8 TableXXXVIII-V24-HLA- A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 Fscore I YVCTPVPHPD 10 L [LTMYVCTPVP| 8 I6 lMYVCTPVPHP 8 9 CTPVPHPDPP| 8 I0 TPVPHPDPPMI 61 ASLTMYVCTP 5 TableXXXVIII-V25-HLA- A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
os] 1234567890 Ilscore RTPTRQISSIII 14 15] PTRQISSIDT 91 I10|SSIDTDPPADII 7 ITRQISSIDTDII 6 TableXXXVIII-V25&26- HLA-A26-10mers-PSCA PCT/US2004/017231 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
o 1234567890 ore ]IRTPTRQISSS 14 []IPTRQISSSDT|I 9 TableXXXVIII-V26-HLA- A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
pos 1234567890 I f RTPTRQIGSS -14 i PTRQIGSSDT 9 TableXXXVIII-V27-HLA- A26-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
PosI 1234567890 Iscore 1 PSRGQALRRAI 4 2 SRGQALRRAQ- 2 TableXXXIX-V1-HLA- B0702-10mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
1Pos 1234567890 Isco 101 QPAAAILALL 23| 108 ALLPALGLLL1I 151 WO 2005/014780 TableX)(XIX-V1 -HLA-1 B0702-1 Omers-PSCA Each peptide is a portion of SEQ I D NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptidle is the start position plus nine., 104 AAILALLPALI 14 106 IFLLALL 14 jj6] LALAGA 13 [12 G1LPGA 131 49 WTRRAG 13 TAFRAOL 1 110 0 LPA LL 1 3 F4 FALMG 12 13 LAQPT L 12 F52 RFLL 12 82 KNTCDD 12 F91 LCSGHA 12 1 03 AAILLLA12 F16 QFilLlY 1 F27 KAVNEC 11 FLVNCQ 11 94 ASGHAQP 11 97 FHLQAAI 11 98 HALPAAI L42 TableXXXIX-V4-HLA- B0702-1 Omers-FSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
PsI 124679 176 SPPSG L 23 8 5 VPA AL 22 102 VPP PML 22 126 PPDGSNL 22 F27 FPSR PL 21 1100 APPPDP [-21 TableXXXIX-V4-HLA- B0702-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1 2345676890 1 66; vPQ-A-S-VPLL-T][ 19 76 HPQPL 19 60 QPLWQ 18 813 GP LCF 18 :48 DPSRF 17 25 GP SRP 16] 178 ARGARR 1 33 PLCLS 14 50 ASFWG 14 64 GWQ P 14 104 HPPPASR 1 :31] RLPLFS 13 54] L WGAPL QP TL F13 75 THAW FL 13 23:1 EAPMC RL[l 81 EDAVEHP 1 14 PASLNV 121 17 TPCTG 11 45 CSGPASYRL 11 F71] VLT 7AW 11 110 AL PTQ 11 F11 IGFTD 11 125 DPAGP 11 11NPLOCFHP 1 PCT/1§S2004/017231 TableXXXIX-V4-HLA- B0702-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine, [Pos 1235689 rL e [148 NPLRLFQ [160 FPHPYDS i Tab~eXXXIX-V1 9-H LA- B0702-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
Po-s 12346789 B0702-1 Omers-PSCAI Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
pos 123567 r0 e 76 DPSRL 17 D SSLW FP 16 [ID SGPASRE1 TableXXXIX-V21 -H A- 1 B0702-1 Omers-PSA WO 2005/014780 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 11234557890 1 1 DPQEPL 19 Fj-DPQW V '13 ASPT F 11 [TableXxXlIX-V21 &22-H LA-i B0702-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nne IPos 12467 scorel F1 QAVPLTJL 1 F2S] LLDL 10 F-9] DLAQWEPVLVI-9 TableX)(XIX-V22-H LA- 20702-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Posil 1234567890 IE F1IQAVLLH 13 LF4 FHAWEV 13 F2 ASVPLLTHLA IF10 TabeXXIX-24-H LA- B721mes-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is9 the start position plus nine.
Pos 124679 F [TableXXXlX-V25-H LA-j Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
F-91T~lSDT7 TableXXXIX-V25&26- HLA-BU702-10mers-
PSCA
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino'acids, and the end position for each peptide is the start position plus nine.
[Posl 1245780 9~ LASSTDP 11 [TableXXXIX-V26-HLA- B0702-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide 217 PCT/1§S2004/017231 is the start position plus nine.
FIGS7]PP 1-11 TPTR-QIGSS-D F Tab~eXXXIX-V27-HLA-1 B0702-1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
Po3s 12345-67890 Fe] Z1 PSRQARR Zo TableXL-V4-HLA-B308- I Oimers-PSCA ,FNoRasultsFound.
1TabeXL-Vl 9-H LA-B08-1 1 Omers-PSCA FIF 235780EREe FNoResultsFound. I [TableXI-21-11-A-B308-] L[NoResultsFound.
[TableXL-V21-HLA-BO8-] F [PLs 1245 80 e LL-NoResultsFound.
~TabeXL-V21 &22-H LA- BOB-I Omers-PSCA [P os 1234567890c oreI L-ofjesutsond WO 2005/014780 [TableXL-V22HLA-B08-] I Omers-PSCA FposI 124679 EcR FNoResultsFound.I [TableXL-V24-HLA-B08-1] 1 Omers-PSOA 11 Li NoResultsFound TableXL-V25-HLA-BO8- 1 Omers-PSCA [o sI 123456890co re [i NoResultsFound.
I
[TableXL-V25&26-HLA- 808-1 Omers-PSCA posI 1235689 Iscorel Li No ResultsFound, STableXL-V26-HLA-B08- I Omers-PSCA o s 1235689 scor e [NoResultsFound TabIeXLI-V1 -H 81510-1 Omers-FC [p-osi 123456789 FNoResultsFound. I F TableXLI-V4-HLA- Ios 135780-10ers-PS BI1-1s OErs-PSCAO N77oResultsFound.
TabieXLI-VI 9-H LA- 81510-1 Omers-PSCA FPosiz 12356790e INoliesulisFound
I,
BI51-1OmrsPSCA FposN 124568901re N77oResultsFoundl.
TableXLI-V21 -HLA- 1 Bi 510-lomers-PSCA FPos 1245689 FNoResultsFound.
ETableXLI-V21 &22-H LA-] 8151 0-l0mers-PSCA IPos 1234567890re Li NoiesultsFoundl.
TableXLI-V22-HLA- [PosI 1235689 [NoResultsFound.
FrTableXLI-V24-HLA- 1 [BI 51 0-l0mers-PSCAJ FNo ResultsFound 1 [BiSI 0-l0mers-PSCA [NoResultsFound. [TableXLI-V25&26-HLA- SB151 0-l0mers-PSCA [Poeg 124679 ERi I NoResultsFound. TableXLI-V26-HLA- 81510-1 Omers-PSCA FPos 124679 ER NoResultsFound.
TableXLII-VI -H LA- B2705-10Oners-PSCA PoeI 12346789 FNoResultsFound. PCT/1§S2004/017231 M oResultsFound.
82705-1 Omers-PSCA] Iposi 1235689 i INoResultsFound.__I TabieXLI I-V21-H LA- 82705-1 Omers-PSCA Posi 123567890 [NoResultsFound.
TableXLII-V21 &22- HLA-B2705-10Omers-
PSCA
[NoResuls ond TableXLII-V22-HLA- B2705-1 Omers-PSCA Pos 1234567890 I NoResultsFound.
[TableXLII-V24-HLA- 1 823705-1 mers-PC [[NoRsui sFound.
STableXLII-V25-HLA- 1 82705-1 Omers-PSCA PE 12346789 [[NoResultsFound.
[TableXLII-V25&26- HL -B20 Omers- [NoResultsFound.
TabeXLII-V26-H LA- B82705-1 Omere-PSCA1 FP-os 12346789 F-oResultsFound. TableXLIII-IHLA- 82709-1 Omers-PSCAl TableXLI -VI 9-H LA- 82705-10Orers-PSCA WO 2005/014780 FPos score89~~ NoResults!Found.
TableXLlIl-V4-HLA- Bps 27890sPC B2709-1 =ore NoResulisFound.
TableXLIlI-V1 9-HLA- 22709-1 Omers-FSCA [~osI 1234567890 No ResultsFound.
22709-1 Omers-PSCA Pos 1234567890re FNoResulisFound.
TableXLI ll-V21 -HLA- B2709-1 Omers-PSCA IPosi 123456789 FNoResultsFound.1 TableXLIlI-V21 &22- HLA-B2709-1 Omers-
FSCA
Iposl 123457890 NoResultsFound.j FTableXLllI-V22-HLA- B~s 27890s-SC 709-1E =ore FNoResultsFound.1 TableXLllI-V24-H LA- B2709-1 Omers-PSCA FPosI 123 s6790 e~ ,FNoResultsFoun TableXLl lI-V25-HLA- B2709-1 Omers-PSCA PosI 1235689 Ls co r NoResultsFound.
FTableXLlll-V25&26-1 HLA-B2709-1 Omers-
PSCA
FNoResultsFound FTa-bleXLIII-V26-HLA- IB2709-1 Omers-PSCA Pos 124679 Ri FNoResultsFound. TableXLIV-VI-HLA-84402- 1 Omers-PSOA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
104AILLPL] 20 108ALPLLL] 19 :97AAQPAI] 5 109LLAGLWI i PGAL I 3 [106 IALAGLI 3 21E9WARRI 2 PCT/1§S2004/017231 TableXLV-V1 -HLA-B4402-~ 1 Omers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
68 LNCD1D 14 ELPGALC 9 44 LGQWTR 9 [TablemLV-HLA-4402- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptidle is the start position plus nine.
469 SGPAYLW 1 159 AFAPYL 17 88-2 AHNFLM 16 110 ALRPRI 16 146 TLP LL 1 17] RLPLRS 156 43 ACSGPSY 1 [50 ASRL PL 1 64 GVPAVL q1 71 EPETPQW 1 80 WEVV E RH 1 16 HIYLS ER1 F23 PA MCRL 1 VVQAVLL 1 11FN TRIS 13 L138 CFGAFT Liq WO 2005/014780 TableXLIV-V4-HLA-B4402- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 score 142 PAFSTLNPVLI 131 151 LRHLFPQEAF 131 THPAQWEPVL 12 86 PEAHPNASLT 121 126 PPADGPSNPLI 12 130 GPSNPLCCCFI 121 135 LCCCFHGPAFI 12 27 MPCSRLPPSL 11 CSGDPASYRLI 11 54 LWGAPLQPTL 11 VPEAHPNASL 10 156 PQEAFPAHPI 101 158 EAFPAHPIYD I 91 TableXLIV-V19-HLA- B4402-10mers-PSCA Each peptde is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 sc I 'AGPMPCSRLLII16 8 RLLPSLRCSL 15 S411MPCSRLLPSLII 11 I 21GPMPCSRLLPI B4402-1 mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 score 4ISGDPASSRLW 171 D IASSRLWGAPL 15 SCSGDPASSRLI 11 TableXLIV-V21-HLA- B4402-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 1re 5[VPLLTDPAQW F14 9 TDPAQWEPVL| 12 3 ASVPLLTDPAI TableXLIV-V21&22-HLA- B4402-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 score 4I VPLLTDLAQWI 14 -1 QASVPLLTDL 121 8 TDLAQWEPVL -12 21 ASVPLLTDLA 8 I\ SVPLLTDLAQ 61 TableXLIV-V22-HLA- B4402-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 10 amino acids, and the end position for each peptide is the start position plus nine.
Ds 1234567890 S|VPLLTHLAQWI 14 lI QASVPLLTHL 12 8 |THLAQWEPVLl 12 2 ASVPLLTHLAI -31 SVPLLTHLAQ 6 PCT/US2004/017231 TableXLIV-V24-HLA- B4402-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
Posl 1234567890 score [211ASLTMYVCTPI1 7 [IINASLTMYVCTI 4 1 LTMYVCTPVPI 3 S71 YVCTPVPHPDI 3 [811VCTPVPHPDPII 3 S10 1TPVPHPDPPMEI 3 B4402-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 [31RTPTRQISSI 13 [10 SSIDTDPPAD|1 8 TableXLIV-V25&26-HLA- B4402-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus nine.
Pos 1234567890 I81SSSDTDPPADI 171 RTPTRQISSS I 51 RQISSSDTDPI 3 611 QISSSDTDPPI TableXLIV-V26-HLA- B4402-10mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each WO 2005/014780 WO 205/04780PCT/1§S2004/017231 start position is specified, the length of peptide is 10 amino acids, and the end Position for each peptidle is the start position plus nine.~ FI]RTPTRQIGSS F- RQIGSSDTDP IF- 79 SSTDPA 4 SDTDP1ADG3 TableXLIV-V27-HLA-1 B4402-1 Omers-PSCAJ Each peptide is a portion of SEQ ID NO: 8: each start position is specified, the length of peptide is 10 amino acids, and the end position for each pepticde is the start position plus nine.
FfTableXLV-V1 -HLA- 01-1 Omers-PSCA [posI 235689 IcoEl FNoResultsFound.j FrTableXLV-V4-HLA- 1 B51 01-1 Omers-PSCAJ STableXLV-V1 9-HLA- 1 B51 01-1 Omers-PSCA] ,FNoResultsFound. lj 1 B5101-1 Omers-PSCA os 1235889 scor Fr oResultsFound._Jj FTableXLV-V21 -HLA-1 85101-1 Omers-PSCA [f 135689scorel NoResultsFound.
TableXLV-V21 &22- HLA-BSI 01-1 Omers-
PSCA
[PosI 1234567890 r~ ,ENoResultsFoud ETableXLV-V22-HLA- [BSI 01 -l0mers-PSCA FNoResultsFound.
ETableXLV-V24-HLA- B5101 -l0mers-PSOA [posii 2346789 [NoResultsFound.
i TableXLV-V25-H LA- 1 B51 01 -lomers-PSCA ~PosI 13456789 FNoResultsFound.
LTableXLV-V25/26-HLA- B51 01 -l0mers-PSCA Pos 12346789 ,FNoResults~ound.
65101 -l0mers-PSCA [PosIF 12346789 FNoResultsFound.
TableXLVI-V1 -HLA-DREI-0101- 1 5mers-PSCA Each peptidle is a portion of SEQ ID NO: 2; each start position is specified, the length of paptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
[P-osl 123456789012345]sor [1041 AAILALLPALGLL ]35 AVLLALLMAOGLALQ-P]f__33 10 MAGLALQPGTALLCY 32 [21 KAVLLALLMAGLLQ ]F 26 [50 TARIRAVGLLTVISK [[25 LALLMAGLALQPGTA]IF 24 LTableXLVI-V1-HLA-DRB1-01 01- 1 Smers-PSCA Each peptide is a portion of SEQ I D NO: 2; each start position is specified, the length of pepide is 15 amino acids, and the end position for each peptide is the I start position plus fourteen.
[Pos 12345678901234-5 Re] 47QCWTARIRAVGLLT 24~ F 88 DTDLCNASGAHALQPF 241 F97 AHALQPAAAILAL7LP [[241 [1061 ILALLPALGLLLWGP 241 jj4] VLLALLMAGLALQG 23 52 RIRAVGLLTVIKGC 2 [55 AVGLLTVISKGCSLN 23A 56l VGLLIVISKGCSLNC 123 95 SGAHALQPAAALA 23 LMAGLALQPGTALLC]22 94l ASGAHALPIA F22 100 LQPAAAILALLG 2 2 103 AAAILALLPALGLL 22 571 GLLTVISKGCSLlNV 861CCDTDLCAS HA 19 [101] QPAAAILALLPA=LGL F19 89 jTDLCNASGAHALQPA 18j 109 LLPALGLLLWGPGQL 1 33 EDCLQVENCTQLGEQ 17 53 IRAVGLLTVISKGCS F17 107 LALLPALGLLLWGPG 17 TableXLVI-V4-HLA-DRBI-0101- 1 Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptidle is the start position plus fourteen.
Posj 12345678024 scor 1148 NPVLRHLFPQEAFPA[ F-321 F151]1 LRHLFPQEAFPAHPI IF 31] 169] SQVWVSVVSPAPSRGQ][F F621 TLGVVPOASVPLLTH ][29] [NJ6 TRQIGSIDTDP D] F281 1165 IYDLSQVWSWVSPAP[ IF281 [681 LSQVWSVVSPAP=SRG 281 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TableXLVI-V4-HLA-DRB1 -0101 -1 I Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptidle is the start position plus fourteen.
[P-os 123456789012345 soe [131 SRAVTPTCATPAGPI][26 TTTWARRTSRATT 251 F48 DPASYRLWGAPLQT]24 58 GAPLQPTLGVVPQAS 1[ 24j 771 PAQWEPVLVPEAHPN] F24 [I11 GPAFSTLNPVLRHLF][ 241 [1621 AHPIYDLSQV VS] F241 ]RRTSRAVTPTAP] F231 54 LWGAPLQFTLPQ [F231 67 PQASVPLLTHPAQE][231 108 IPMALSRTPTRIS] F23 1144 FSTLNPVLRI-LPE] 3 F49 PASYRLWGAP LQPL[ 2 QPTLGVVPQASVPLL[ F221 821 PVLVFEAHPNAST ]{22 F92 ASLTMYVCAPPP ]E[22 F99 CAPVPHPDPPM ALS 2 F59 LQPTLGVVPASP[ 201 F94 LTMYVCAPVPHPP] Eol [120 GSIDTDPPADGPSNP[ 19 1163 HPIYDLSQVWSWP[ 7 i TWARRTSIRAV ETA 11 52 1YRLWGAPLQPTVI [718 1 91[NASLTMYVCAPVPHP[ 1 18 1361CCCFHGPAFSTLNPV 1-8] [157 QEFA-PY Q F181 AVTPTCATPAGP-MPC][- 171 171 [61] PTLGVVPQASVPLT] 17 [681 QASVPLLTHPAQE[ 171 [71] VPLLTHPAQWEPVLV[ IF171 WVEPVLVPEAHPNFL[[17 [81 EPVLVPEAHPNASLT [171 [1-72 WSVVSPAPSRGQALR[ F171 [16 VTPTCATPAGPMCSF[-1 [17TPTCATPAGPM PCSR I[:16 TableXLVI-V4-HLA-DRBI -0101 1 5mers-PC Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptidle is the start position plus fourteen.
[Pos 12345678901234 ~7 221 TIPAGPMPCRLPS LI 24 AGPMPCSRLPPS-LRC 1- 291 CSRLPPSLRCSL-HS-A 1- 1 39 SLHSACCSGDPASYRF 16 63@ LGVVPQASVPLLTH ]16 701 SVPLLTHPAQWEPVL IF 16 I 79 QWEPVLVPEAHPNASII 6 [1041HPDPPMALSRTPTRQ[ 1-s [132 SNPLCCCFHGPAFST -16 1-33 NPLCCCFHGPAFSTL]IF 16 [134 PLCCCFHGASL] 1-6 145 STLNPVLRHLFP ]1-6 154 LFPQEAFPAHID 1[16 1I74VSARGQAILRRAI[ 16 S31 IRLPPSLRCSLHSACC][r15 [g53 RLWGAPLPLG P[5 LI LTHPAQWEPVLVPEA] 15 87EEAH PNASLTNYVCAP] []15 [1os6] IDPPMVAILSRTPTRIG][is5 F11 SIRTIPTRIDP ][15 1113 RTIPTRQGITP 5i 1123I DTDPPADGPSNPLCC][ is5 1171 IVWSVVSPAPSRGQAL][I TableXLVI-VI 9-HLA-DRB1 -0101- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 16 amino acids, and the end position for each peptide is the start position plus fourteen.
[Posl 12345678901234 e I MPCSRLLPSLRSLH 17 [D TPAGPMPCRLPS 16 I AGPMPCSRLPLR 161 I iCSRLLPSLRCSLHSA 161 12[SRLLPSLRCSLHSAC 16! TableXLVI-VI 9-HLA-DRB1-0101-1 1 Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
Pos I1234567890123457,Fc-oe] 13 RLLPSLRCSLHSACC ::3ATPAGPMPC;SRLLPSE~jq :81PMPCSRLLPSLRCSL .1 :10 PCSRLLPSLRCSLHS EA 1j6] LPSLRCSLHSACCSG 71]TCATPAGPMPCSRLL Z81 :5]PAGIPMPCSIRLLPL l =7 GPMPCSRLLPSLRCSi 1 14I~ LLFSLRCSLHSAC [:I:A TableXLVI-V20-HLA-DRBI -0101- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
Posl 1234567890124 ~e1 1 DPASSRLWGAPLP 124 75[ SRLWGAPLQPTLGV 8 j2] SLHSACCSGDPAS 16 =1]CSLHSACCSGDPASSI 1- 4]HSACCSGIDPAL =1 ACCSGDPASSRLWGA F 13 ASSRLWGAPLQPTL1 4 12FP-ASSRLWGAPLQPTL =1 FTablexLvl-V21 -HLA-DRB1 -01 01-' Each peptide is a portion of SEQ ID NO: 8: each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
[Pos 1234567890123-45 E.Fs-oeI [I TLGVVPQASVPLLTD]DJ [Aj PQASVPLLTD PAQWEEA~ WO 2005/014780 WO 205/04780PCT/1§S2004/017231 IIQASVPLLTDPAQE 17z VPLLTDPAQWEPVLV :2 F-2]1 LGWVPQASVPLLTD FZ1 6 F9]1 SVPLLTDPAQWEPVL 71 F13 LTDPAQWVEPVLVPEA 5 F121 LILTDIPAQWEFVILVIPEf F1 TableXLVI-V21 &22-HLA-DRB1 010l-lflmers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end positio ,n for each peptidle is the -start position plus fourteen.
Pos 11234567890123457 score ng~ IVPLLTDLAQWEPVLV] 26 PQASVPLLTDLAW L2 [12 LTDLAQWEPVLVPEA 23~ F-6 QASVPLLDAQE 17 F-1 LGVVPQASVPLLTD 16 F-8]SVHLLTIJLAQWEPVL 716 11E LLTDLAQWEPV=LVPE 14I Tab~eXLVI-2HLA-DRBI -01 01-] 1 mr-PSCA Each peptidle is a portion of SEQ ID NO; 8; each start position is specified, the length of peptidle is amino acids, and the end position for each peptidle is the Istart position plus fourteen. I [Pos 1 123456789012345 1 scor [j9] VPLLTHLAQWEPL L-25 LAQWEFVLVPEAHPN 241 PQASVPLLTHLA E 231 [12 LTHLAQWEPVLVPEAF 23] F-]QASVPLLTH-LAQWEP 17 71LGVVPQASVPLLH 16 Ft81 SVPLLTHLAQWEPVL 16 []ii LLTHLAQWVEPVLP F14 TableXLVl-V24-LA-DR11-0101- 1 5mers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start pcsition is specified, the length of peptidle is 15 amino acids, and the end position for each peptide is the start position ptus fourteen.
[Pos 1234567891245 j~je 1 4 CTPVPHPDPPMALSR j-22 [3IJASLTMYVCTPVP-HPD 720 F-]NASLTMYVCPVH 19 79 LTMYVCTPVPH FP 1 9 D EAHPNASLTMYVCTP 15 11 jMYVCTPVPHDPM 15 E PNASLTMYVCTVP 14 ISLTMYVCTPV D F14 110 T VTVHDP 14] 121 YVCTPVPHFDPPMAL [F 141 15 TPVPHPDPPMALSRT I[ iM TableXLVI-V25-HLA-DRB1 -01 01- 1 Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of paptide is amino acids, and the end position for each peptide is the start position plus fourteen.
[Posj 12345678901234 [scores [11 ]TRQISSIDTDPAG[-28, [3i1 PMVALSIRTPTRQISS 23 15 jSSIDTDPPADGPSNP 19:j F- ]DPPMALSRTPTRQIS][15j D IRTPTRQISSDTP 14 F14 ISSIDTD)PPADGSN 14 ID SIRTPTRQISSIOTDP 13 TableXLVI-V25&26-HLA-DRBI- 0 101-1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptide is the start position plus fourteen, Posj 12345678901234 EFE 11 MASTTRQISSS I 2- I q TRQISSSDTDPPADG [L22 13 SSSDTIDPPAD-GPSNP 19 D RTPTRQISSSDTDPP -1-4 DI SRTPTRQISSSDTDP 13q TableXLVI-V26-HLA-DRBI -0101- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptidle is the start position plus fourteen.
[Posl 123456789012345s-core [DIi PMALSRTPTRQIGSS] 2[ [I9I TRQIGSSDD FAD 221 [13 JGSSDTDlPPADGPSNP 19N L-5] SRTPTRQIGSSDTDP[ 151 [-Sj RTPTRQIGSSDTP] 1T51 I[AjALSRTPTRQGSST 11 beLI-V27-HLA-DRBI-01 01- [TaleXV Sers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptidle is the start position plus fourteen.
Pos 123456789012345 scorej [lIi SPAPSRQQALRRA L8 TabeXLVII-V1-HLA-DR81-0301-1 I Each peptidle is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptidle is the star position plus fouren P08 123456789012345 scorel 67 SLNCVDDSQDYYVGK F261 106 ILALLPALGLLLG 24] KAVLLALLMAGLALQ] 22 F10 IMAGLALQPGTALLCY[ I211 104 AAILALLPALGLL F21] 66 jCSLNCVDDSQDYYVG][ 19j 33 EDCLQVENCTQGEQIF181 56 VGLLTVISKGCSN 181 [19 TALLCYSCKAQVSNE]I 17 135 CLQVENCTQILGEC 17A WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TableXLVII-V1 -HLA-IJRB1-0301-1 1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start I position plus fourteen.
PosIl 123456789012345 Ii LLALLMAGLALQPT1 F 1 MKAVLLALLMAGLAL 16 1031 AAAILALLPALGLLL 15 F18 GTALLCYSGKAQV-SN 1 F251 SCKAQVSNEDCLQVE 14 [58 LLTVISKGCSLNV 14 F-41 VILLALLMAGLALP 13 111AGLALQPGTALLY 13] F-12 GLALQPGTALLCYC[3 TARIRAVGLLTVS 134§ 841 ITCCDTDLCNASGAHJ 131 [107 LALLPALGLLLWP 1[13 F-3] AVILLALLIAAGILAP 1[ 12 61~ LALLMAGLALQPGTA 1!121 F281AQVSNEDCLQVENTJ 12] [41] CTQLGEQCWTA EIA12 556] AVGLLTVISKGCSLN 1[ 12 59 LTVISKGCSLNCVDD![ 121 F81 KKNITCCDTDLCNAS 12 F88 DTDLCNASGAHAQ] 121: F97 AHALQPAAAILAL 1112! [1021 PAAAILALLPALL 11 12 los ILALPAGLLWG 11 12! {TableXLVII.V4-HLA-DRBI-0301-] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.- [Posl 123456789012345 [144 FSTLNPVLRHLFPQE 27I [29 CSRLPPSLRCSLHSA7d [119] IGSIDTIDPPADGPS 22 [62 TLGVVPQASVPLLTH F .21 [iooAPVPHPDPPMA=LSRT L21 TableXLVII-V4-HLA-DRBI-0301-1 1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the Istart position plus fourteen.
Pos 1234567890123457 [coe 6LGWPQAVLLH[ 20 60QPTLGVVPQASVFLL[[ 181 162 AHPIYDLSQVWSVVS[ r 18 F71 VPLLTHPAQWEPVLV[ F 149 PVLRHLFPQEAFPAH[[ 17~ 1571 QEAFPAHPIYDLSQV[ 17 L 831 VLVPEAHPNASLTMY L161 136 CCCFHGPAFSTLP 76 [1401j HGPAFSLPRH rl 6 L-51]YRLWGAPLQPTLGVV [15 F-76 SVPL-LTHPAQWEPVL 14 E[73 LLTHPAQWEPVLVPE 14 [33 PPSLRCSLHSACC1SG [56 GAPLQPTLGVVPQASF 13 [80 WEPVLVPEAHPNASL[ 131 581 EPVLVPEAHPNASLT] 13 [82 PVLVPEAHPNASLTM 13~ [fj NASLTMYVCAPPH 73 [4J LNPVLRHLFPQE F 13] [161l PAHPIYDLSQVWSVVF7 j TableXLVII-V1 9-HLA-DRI-0301-1 I Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
Posj 123456789012345 5cr II CSRLILPSLRCSHF 28 I ~LPSLRCSLHSACCSG L 1 3] STableXLVI -V20-HLA-DR -0301 I 5mers-IPSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
[P05 1 123456789012345 soe [1Is SRLWGAPLQPTL-GVV [14 SSRLWGAPLQPTG Zi17[ [5]ISACCSGDPASSRLWG Ill [ii IDPASSRLWALP L19[ I 7] ACCSGDPASSRLWGAIZa! [D CSGDPASSRLWGAPLIIA TableXLVII-V21-HLA-DRI -0301- 1 Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the -start position plus fourteen.
Posj 123456789012345 [EjjR [D SVPLLTDPAQWEPVL][ 24 DjI TLGVVPQASVPLLTD]F 271 F-2]1 LGWVPQASVPLLTDP] 201I [10 VPLLTDPAQWEPVLV 17i E12LLTDPAQWEPVLVPEE LII [jQASVPLLTDPAQWEP L:jAI TableXLVll-V21 &22-HLA-DRI 0301-1 Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the Istart position plus fourteen.
I~s123456789012345 .[coe [8 SVPLLTDLAQWEPVL 241 [I]ILGVVPQASVPLLTDL] 201 [D VPLLTDLAQWEPVLV 171 [EIiLLTDLAQWEPVLVE 1 61 [D QASVPLLTDLAQWEP] 13 12LTDLAQWEPVLVPE:A II131 TableXLVI l-V22-HLA-DRI -0301- 1 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Each peplide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the Istart position plus fourteen.
[Pos 11234567890123457 F50- FJ LGVVPQASPLLHL 1201 79 VPLLTHLAQWEPVLV 171 11 ILLTHLAQWEPVLVPEl 161 F 1 SVPLL-THLAQWEPV L_ 14 76 QCASVPLLTHLAQWEP 1 3 1-2] LTHLAQWEPVLVPEA 13 D VPQASVPLLTHLAQW LI TableXLVII-V24-HLA-DRI-0301- I fmers-PSCA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
Pos 1234567890124 [orel~ 6]TPVPHPDPPMALSRT 21 j-6] NASLTMYVCTPVH 12A j8 SLTMYVCTPVPHPD 11, [jj]0 TMYVCTPVFHPDP F-1-0 14 jCTPVPHPDPPMALSRE101 TableXLVII-V25-HLA-DRI-0301- 1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
F14 ISSIDTDPPADPS F22 DP LS RTPTRQI S 111 PMALSRTPTRQISSI ll- F-1-1 TRQCAS S ID T DPPADG] FI 1] [12 RQISSIDTDPPADCP [ot TableXLVII-V25-HLA-DRI-0301 I 5mers-PSCA
I
Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen. I [Pos 123456789012345 scoe] [Z1 PMALSRTPTRQISSS Li11] [D TRQISSSDTDPPADG 11 RQISSSDTDPAG II] 72 MALSRTFTRQISSSD]F[ 6 I ALSRTPTRQISSSDT [TableXLVII-V26-HLA-DR1-0301] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the -start position plus fourteen.
[P05 123456789012345 scorel [12 IGSSDTDPPADGPSN[[ 12] F-1]1 PMALSRTPTRQIGSSI[ 11] [D7ITRQIGSSDTDPPD I1l [10 RQIGSSDTPAG il1l MALSRTPTRQIGSSD F78 [D ALSRTPTRQIGSSDT F 8] TableXLVII-V27-HLA-DRI-0301- 1 Smers-PSCA Each peptide is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end postion for each peptide is the start position plus fourteen.
Pos 123456789012345 JFEe [:]VVSPAPSRQQAL RRA 17 7] VSFAFSRQQALRRAQj 9I Tab~eXLVIII-V1-HLA-DR1-0401- Each peptide is a portion of SEQ IC NO: 2; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the star position plus fourteen.
E os 123456789012345 soe D7] KAVLLALLMAGLALQ]I D7] AVLLALLMAGLALQP].- N61 LLALLMGLQT 27 JKAQVSNEDCLQVN] F201 [33~ EDLVN LE Q]F 201 F50 TARIRAVGLLTVISK ]F201 531 IRAVGLLTVISGC F55 1AVGLLTVISKOSL] 56~ VGLLTVISKGCSLNC][ 201 8DTDLCNASGAHALQP][ 97AHALQPAAAILALL [104 AAILALLPALGLL [1061 ILALLPALGLLG [71 LLMAGLALQPTA] 18 32 E D CLQVENCTQLE][F 18 52RIRAVGLLTVISKGC] 18 93 NASGAHALQPAAAIL][ 18 21l LLCYSCKAQVSNEDC] IF17 74 SQDYYVGKKNITCCD[ F186 7]MKAVLLALLMAGLAL][F 14 Li[ALLMAGLALQPGTAL 141 10~ MAGLALGTLY[ 14 18 GTALLCYSCKAQVSN[ 141 19j TALLCYSCKAQVSNE][ 14 35 NCLQVENCTQLGEQC 14 F59 LTVISKGCSLNVD Zi14] L 65 GCSLNCVDDSQDY F 14 [68 LNCVDDSQDYYVKK 14 [81l KKNITCCDTDLCNAS][ 14 103 AAAILALLPALGLLL 4 107 LALLPALGLLLWGP][14 [61 LMAGLALQPGTALLC] 12 [ii AGLALQPGTALLGYS][ 12 [14 ALQPGTALLCYSKA[ 121 [16 QPGTALLCYSCKAQV][ 12 171I PGTALLCYSOKAQVS I 12 LA YSCKAQVSNEDCLQV][ 12 [29 QVSNEDCLQVENCTQ][12 3911 ENCOQLGEQCWVTARI F4011 NCTQLGEQCWTARIR I 42] TQLGEQCWTARIRAV]12 F44 LGEQCWTARI FL~ 12 45 GEQCWTARIRAVGL][12 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TableXLVII I-V1-HLA-DR1 -0401 Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
7Pos 123456789012345 soe 471 QCWTARIRAVGLLTV I 2 49 WTARIRAVGLLTVI [121 -661 CSLNCVDDSQDYVI 121 67 SLNCVDDSQDYYVGKI[ 12 [701CVDDSQDJYYVGKN [1-2] [73 DSQDYYVGKKNIC [-12 [781 YVGKKNITCCDTL 121 [821KNITCCDTDLCNASG IF-121 [841 ITCCDTDLCNAGA J[ 12 TCCDTDLCNASAHJ 12 DLCNASGAHALQP[121 [94 ASGAHALQPAALJ 2 F98 HALQPAAAILALP [I 121 99 ALQPAAAILALLA [I 12 [101 QPAAAILALLPALLI 121 [102 PAAAILALLPALL 11 12 [46 FQWTRRAGLT[ 1 QDYYVGKKNITCCDTj I 1l [58l LLTVISKGCSLN=CVD [I ]9 [Tab~eXLVIII-V4-HLA-DRI-0401- Each peptide is a portion of SEQ 1D NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptidle is the start position plus fourteen.
[Pos F123456789012345 se QPTLGVVPQASVL 1126 F 681 QASVPLLTHPAQEJ 261 [81] EPVLVPEAHPAL 1126 [162 1AHFIYDLSQVSV 1126 1651 IYDLSQVINSVVAP1 26 [172 WSVVSPAPSRGQALR 261 [52 YRLWVGAPLQPTLGVV 1=2 [77 PAQWEPVLVPEAP [22 [16 SQVYWSVVSPAPSG j tTableXLVIII-V4-HLA-DR1-0401 l5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
[Posj 123456789024 F29 CSRLPPSLRCSLS 20] F511 SYRLWGAPLQPTLGV IF201 S62jTLGWVPQASVLT ]i20 [I3LGWVPQASVPLLH [120] 82 PVLVPEAHPNASLTM 11 20 1108 PMALSRTPTRQIGSI ]F201 116 TIRQIGSIDTDP PADG ]I 201 132 SNPLCCCFHGPAF] 01 14FSTLNPVLRHLFPQE F 201 1148 NPVLRHLFPQEAFPA]I 20] 1681 LSQVWSVVSPAR] F20 67POASVPLLTHPAQWE 105 PDPPMALSRTPTRQI ]IF 181 1131 RTPTRQIGSIDTDPP] 181 [1371 CCFHGPAFSTLNPV FJ 181 [jA TTTWARRTSRATP] 17 TableXLVIII-VA-HLA-DRI -0401-1 1 Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen. Ps123456789012345 E STTWARRTSRAVTPTC 12 I ARRTSR TPAT F12 10 RRTSRAVTPTCATPA [12 11 RTSRAVTPTCATPAG F12 [I PSLRCSLHSACCSGD 12 [43ACCSGDPASYRLWGA FZ12] [48 DPASYRLWGAPLQPT LII2 54 LWALQT VPQ 121:j 57j APLQPTLGVVPQASV[ 1121 59q LQ)PTLGVVPQASVPL II 12] F721 PLLTHFAQWEPVLVP]11 12A 83g VLVPEAHPNASLTMY 11 l2A 85 VPEAHPNASLTMFCJ1121.
87 EAPASTYCAIP] 12 96 IMYVCAPVPHPDPPMA]I 12 1100 IAPVPHPDPPMA LSR]I 12 1104 1HPDPPMALSRTPTRQ]I 121 110 ALSRTTRQIGSIDT 2 1117j RQIGSIDTDPPAG II121 1122] IDPAGSPLC ]l 121 [124 TDPPADGPSNPLCCG] 121A F11381 FGF TV-L F121 11461 HGPAFSTLNFVLRHL[12 11451 STLNPVLRHLFPQEA][ 121 149 IPVLRHLFPQEA F 121 154 LFPQEAFPAHPY F][121 159 AFPAHPIYDLSQVWS[ IF121 1161 PAHPIYDLSQVWV] 121 LL173 SVVSPAPSRGQALR 21 [TableXLI-V19-1-LA-DR11-0401-] Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of poptide is 15 amino acids, and the end position for each peptide is the -start position plus fourteen.
[491 PASYRLWGPQT 16 E 94 LTMYVCAPVPHPDPPIF161 136 CCCFHGPASTNPF 16 141] GPAFSTLNPV rHL] 161 [152j RHLFPQEAFPAHI] 161 [157 QEAFPAHPIYDLSQV[ J161 [1631 HPIYDLSQVWS 16 [jA SRAVTPTCATPAGPM 14 D24lI AGPMPCSRLPPSLRC F[ 141 [33 PPSLRCSLHSCS I 14 [37 RCSLHSAOOSDPSI 14 17]11 VPLLTHPWPL F[ 14 F80 WEPVLVPEAHPNASI14 [91] NASLTMYVCAPVPHP[ 14 99 CAPVPHPDPPMALSR}[ 14 [106] DPFMALSRTPTRQIG3 [14 111 IGSIDTDPPAGS =1 14 [151] LRHLFPQEAFPAHPI ]14 1 THRTTTWARRTSRFV12 [I HRTTTWARRTSRAV/T i WO 2005/014780 WO 205/04780PCT/1§S2004/017231 [PosIj 1234567890124 scor CSRLLPSLRCSLS 20~ [61AGPMPCSRLLPKSLRC 17 F121S-RLLPSLRCSLHSAO 1 141 LPSLRCSLHSACS 14 [--4]TPAGPMPGSRLLPSL] 12 MPOSRLLPSLRCSLH 12 TableXLVIII-V20-HLA-DRI-0401 1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
Posj 1234567890124 k e1 SRLWGAPLQPTLGVV 221 14 S S RLWGA P LQ PTLG-V F201 761 ACCSGDPASSRLWGAI 12 7 ICCSGDPASSRLWGAP F 1 Iii DPASSRLWGAP=LQPT 12 TableXLVII I-V21-HLA-DR1 -0401 I Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the -start position plus fourteen.
[Posj 123458789012345 Ej Ve TLGVVPQASVPLLTD ]_20[ [ZI LGWVPQASVPLLTDP -2]0 [II jQASVPLLTDPAQWEP F20 PQASVPLLTDPAE FL18 F9] SVPLLTDPAQWEP-VL[-j4 Ho 0]VPLLTDPAQWEPVLV Iliii Ta b eXLVI I I-V21 &22- HLA-0DR I- 0401-1 Smers-PSCA Each peptidle is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the s tart position plus fourteen.
[Posl 123456789012345 E TaleL Il-V21 &22-HLA-DRI- 0401-1 Smers-PSCA Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
[Pos 123456789012345] soe F-1] LGWPQASPLD LI20 I I61 QASVPLLTDLQE 20] I JVPLLTDLAQWEPVLV [j PQASVPLLTDLAQWE L18] ISVPLLTDLQEV F14 [121 LTDLAQWEPVVE F14] 1101 PLLTDLAQWEP=VLVP Li2] ITab~eXLViII-V22-H-LA-DRI-0401- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptide is the start position plus fourteen..
[Posl 123456789135 soe [D QASVPLLTHLQE 26] isiLAQWEPVL AP F22 LGVVPQASVPLTH 20 [A VPLLTHLAQWEPVLVF20 75[ PQASVPLTH QE F18 12 LTHLAQWEPLVE 14 10q PLLTHLAQWEPVLVP IF12 TableXLVlll-V24-HL11A-DRI -0401- 1 Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of paptide is amino acids, and the end position for each peptidle is the start position plus fourteen.- Posl 123456789012345 F~o1 6] NASLTMYVCTPVH F14] 14 CTPVPHPDPPMALSR4 721 EAHPNASLTMYVCP 1 ASLTMYVCTPVPHPDI I12] 11]MYVCTPVPHPDPPMAI-12] TableXLVlI-V24-HLA-DR1 -0401- I Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptidle is the start position plus fourteen.
[Posl 123456789012345 [51 TPVPHPDPPMLSRT 12 D7I LTMYVCTPVPHPDPP] 1101 [7q SILTMVYVCTPVPHPD F7B F[10 TMYVCTPVPHPDMI SIB 1 PEAHPNASLTMYVCT][ sI AHNSTYCPVI F- 81 [jjj] HPNASLTMYVCTPVPII 81 13 jVCIPVPHPDPPMALS L I TableXLVIII-V25-HLA-DR1 -0401- Each peptidle is a portion of SEQ I D NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start posiion plus fourteen.
Pos 123456789012345] score I 31PMALSRTPTRIS 201 [i TRQISSIDTDPPADG] 201 DI1 RTPTRQISSIDTDPP]F 181 [E ]IPPMALSRTPTRQIS 141 F14 ISSIDTDPPADGPN] 14] DI~JMALSRTPTRIS F] 121 F-5] ALSRTPTRQISSIDT][21 I112 jRQISSDTDPPAKDGP[ 2 TableXLVIII-V25&26-HLA-DRI -1 0401-1 Smers-PSOA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
FPos 1123458789012345 1 C-]e I PMALSRTPTRQISSS[ I RTPTROISSSDTDPP E 173 I TRQISSSDTDPPADGII WO 2005/014780 WO 205/04780PCT/1§S2004/017231 TableXLVIII-V25&26--LA-DRI 0401-1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide ]is the start position plus fou rteen.
Posl 123456789012345 EI 72 MALSRTPTRQISSSD 73 ALSRTPTRQISSSFT 12] [:11 TableXLVIII-V26-HLA-DR1 -0401- I Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start Position plus fourteen.
3os 123456789012345 scoe] 1] PMALSRTPTRQIGSS 120 6] RTPTRQIGSSDTDPP[ 7j 9] TRQIGSSDTDPPADGI 71 jjALSRTPTRQIGSSDT 17 RQIGSSDTIDPPAFG172 7 TableXLVII l-V27-HLA-DR1 -0401- I Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
3osl 1234567890124 Ede -E jVVSPAPSRQQALRRA La: TableXLIX-VI-HLA-DRBlI1 101- I Smars-PSCA Each peptide is a portion of SEQ I D NO: 2; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
[Pos 123456789012345 ]F E E6 VGLLTVISKGCS=LNC =Zi Tabl~eXLIX-VI-HLA-DRB1 -1 101-1 1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 2; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
FosI 12345678024 46 EQCWTARIRAVGLLT IF27 SQDYYVG-KKN ITCC D[ 20 D I AVLLALLMAGLAL-Q-P 19 1I03 AAAILALLPALGLLL IF 19 E ALLMAGLALQPGTAL[F 18 F44 LGEQCW-T-ARI RAVGL 16 19 TALLCYSCKAQVSNEIF174 73 DSQDYYVGKKNICC 4 85 TCCDTDLCNASGAHAEI-1 rTabIeXLIX-V4-HLA-DRB1-1101- Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 16 amino acids, and the end position for each peptide is the start position plus fourteen.
[ps12345-678901-2345] scorel [169 SQVWSVVSPAPRGQ23 [77 PAQWEPVLVPEAHPN 221 [144 FSTLNPVLRHFQ] 211 [145 STLNPVLRHLFPQEA][ 21 [68 QASVPLLTHPAQWEP I-20! [81 EPVLVPEAHPNASLT [98lIYVCAPVPHPPM Ei2-0 [132 SNPLCCCFHGAS EA [168 LSQVWSVVSPAPSRG]201 [165j IYDLSQ.VWSVVSA[ 19] [l11 TRQIGSIDTDPPG[ 18] [1481 NPVLRHLFPE FPA 1 181 162j AHPIYDLSQVSV] 8 j-2] THRTTTWVARRT-SRAVI 71i 1I41] GPAFSTLNPVLRHLFI[171 I 94 LTMYVCAPVPHPDPP[ 1-6] 11091 MALSRTPIRISI] 151 [13j SRAVTPTCATPAGPM IL 1-4 [23 PAGPMPCSRLPPSLR IL 14 TableXLIX-V4-HLA-DRB31-1 101- Each peptide is a porion of SEQ ID NO: 8; each start position is specified, the length of pepide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
[Ps 12345678901235 soe [291 CSRLPPSLRCSLHSA][ 141 [7j PPSLRCSLHSACCSG][ 141 [45 CSGDPSYL AP] 1-I41 [59 LQPTLGVVPQ)ASVPL[ 1[--4 F71 VPLLTHPAQWEPVLV[ IF141 [105 PDPPMALSRTPTRQI][ 141 [171VWSVVSPAPSRGQA 4 [173 SVVSPAPSRGQALRR][ I4 F53 RLWGAPLQPTL-GVVP] 13 56 GAPLQPTLGVVPQAS] IF131 60 QPTLGVVPQA-sv-PLL][ 131 92 ASLTMYVCAPVPHPD]I[ 131 [151 LRHLFPQEAFPAHPI IL 131 [10 RRTSIRAVTPTCATIPA]L 12 24 1AGPMPCSRLPPSLC] 12 67 PQASVPLLTH PAQWEJ[F 12 79 QWEPVLVPEAHPNS][12 F82 PVLVFEAHPNALT] 121 901 PNASLTMYVCAPVH][121 [99 CAPVPHPDPPM S] 121 [1191 IGSIDTDPPADGPS-N][ 12 TTWARTRATP] 11i TbXLX-V19-HLA-DRRI-111 I 5mers-PSCA 7 Each peptide is a Portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the start position plus fourteen.
I~s 123456789012346 IFZe LIIPAGMP SLR 1 [i C1] SRLLPSLRCS-LHSA] 1-4 [is LPSLRCSLHSACCSG E4i E12 SRLSLCLSAC] 13i III AGPMPCSRLLPSLRCj121 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Tab~eXLIX-V1 9-HLA-DRBI1-1 101- 1 Smers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the -start position plus fourteen, [Posl 123456789012345 je] [3 ATPAGPMPCSRL-LPS]ZIP] [I9] MPCSRLLPSLRCSLH 7: L 8] PMPCSRLLPSLRS[12 LHS][7]fl -1 101 1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
[Po s 1234567890123-45 ~E1 [31 ICSGDPASSRLWGAPL 4I SRLWGAPLQPTLV 310 jjj[CSLHSACCSGDPASS 6 [321 SLHSACCSGDPAS 16 j-3] LHSACCSGDPSR F-4] HSACCSGDPASSL 776 F-]9 SGDPASSRLWGAPLQ6 Fl1C1 GDPASSRLWG-APLQP 111 iii ]DPASSRLWGAPLQPT[1 [1-3 ASSRLWGAPLQPL 1161 [14 SSRLWGAPLQPTL-G-V][11 TableXLIX-V20-HLA-DRB1 -1101- I 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the start position plus fourteen.
[Posl 123456789012345
]VPLLTDPAQWEPVLVIF-
[31 PQASVPLLTDPAQW-VE 7T TableXLIX-V20-HLA-DRBI-1 101-1 1 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is '15 amino acids, and the end position for each peptide is the I start position plus fourteen.
[Pos 123456789012345 rs'c~] [1IjQASVPLLTDPAQWEPI. 12 D I LGVVFQASVPLLTDP [7: Li VPQASVPLLTDPAQW [7 [141TDPAOWEPVLVPEAHI[7 [Iii TLGVVPQA LT 116 131 IWVPQASVPLLTOA [76 [31 SVPLLTDPAQWEPVL J76 Tab~eXLIX-V21 &22-HLA-DRBI-1 1 101-l6mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the -start position plus fourteen.
[PoI 1345789145 cr [AI]VPLLTDLAQWEPVLVF20 [31PQASVPLLTDLA-QWE 13j [31QASVPLLTDLAQ-WEPF 12, TableXLIX-V22-HLA-DRB1-1 101-1 I 5mers-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is amino acids, and the end position for each peptide is the satpsition plus fourteen.
[Posl 12345678901234-5 coe [311LAQWEPVLEAP [F 27 [31QASVPLLTHLAWE [31VPLLTHLAQWEPVLV [IEIo [31j PQASVPLLTHLAQWVE [I173 ITableXLIX-V24-HLA-DRBl -1101- I Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peptide is the I start position plus fourteen.
[Pos 123456789012345 Iscore I ii IMYVCTPVPHPDPM F-72 1391 LTMYVCTPVPHPP 16 3-71 AS LT M YVCT P vPH PD 3~ [351 PNASLTMYVCTPVPH[[F 12 [1l4 CTPVPIPDFPMALSR12 D31 NASLTMYVCTPVPHP [7 [TableXLlX -V25-HLA-DRB1 -1101-1 15mners-PSCA Each peptide is a portion of SEQ ID NO: 8; each start position is specified, the length of peptide is 15 amino acids, and the end position for each peplide is the Istart position plus fourteen.
[P-osIl 123456789012345 7seor] F11] TRQISSIDTDPPD 18 n49I MALSRTPTRQISSID 141 ISSIDTIDPPADGPSN 121 ALSRTPTRQISSI F- 81 I 151 SSIDTDPPADGPSNPEp13 TbeLX-V25&26-HLA-DRBI- IableXLI Each peptidle is a portion of SEQ ID NO: 8; each start position is specified, the length of peptidle is 15 amino acids, and the end position for each peptide is the Psstart position plus fourteen.
Ej123456789012345]se] [DI MALSRT-PTR-QISSSD ZD TRQISSSDTDPPADG [-12] DI1 ALSRTPTRQISSSDT I 8 13 ISSSIDTPPAIDGPDSNP [1 131l PMALSRTPTRQISSSJ 7]I1 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Table 1: Protein Characteristics of PSCA v.4 PSCA v.4 Bioinformatic URL Outcome Program ORE ORE finder 570 lip Protein length 189aa Tnsieinbrane region TM Pred. http:i/www.ch.embnct.org/ no TM HMMTop http://www.enzimn.huqmumtop/ no TM Sosni http://www.genomead.jp/SOSui/ soluble TMHMM http://www.cbs.dtu.dk/service s/TMHM\M no TM Signal Peptide Signal P http://wwiw.cbs.dtu.dk/servi=e/SignalP/ none p1 p1/MW toot http://www.expasy.ch/tools/ p 1 8.87 Molecular weight p1/MW tool http://www.expasy.cbi/tools/ 20.3kDa Localization PSORT http;//psort.nibb.acjp/ 90% maitochondria PSORT II http://psort.nibb.ac.jp/ 78% mitochondria Motifs Pfam http://www.sanger.ac.uk/Pfainf no motif Prints http://www.biochem.ucl.ac.uk' eadherin signature Blocks http://www.blocks.fhcorg/ Granulin PSCA v.1 Bioinformatic URL Outcome Program ORE ORF finder 372 bp Protein length 123aa Transinembine region TM Fred http://www.ch.embliet.org/ 1 TM, as 99-118 HMMTop http://www.enzim.hu/hmmtop/ ITM, as 103-121 Sosui http://www.geiioiie.ad.jp/SOSui/ membrane protein aa t100- 122 TMHMM http://www.cbs.dtu.dk/services/TMHMM no TM Signal Peptide Signal P lhttp://www.cbs.dtu.dk/servics/SignalP/ yes, aa 1-15 PI p1/MW tool http://www.expasy.ch/tools/ pI 5.01 Molecular weight p1/MW tool http://www.expasy.ch/tools/ 12.9 kDa Localization PSORT http:I/psort.nibb.ac.jp/ 9 1% plasma membrane PSORT 11 http;//psort.nibb.ac.jp/ 34% plasma membrane, 34% extracellular Motifs Plfsm http://www.aangcr.aecuk/Pfam/ uPAR, Ly-6 Prints http://www.biochem.ucl.ac.uk/ no motif Blocks littp://www.blocks,thcrc.org/ Ly-6 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Table LI: Exon boundaries of transcript PSCAv.1 Exon Number Start End Length 1 10 69 2 70 177 108 3 178 985 808 Table 1.I1(a). Nucleotide sequence of transcript variant PSCAv.2 (SEQ ID NO:6527 tttgaggcca ggctgtgctg gtgctactcc ccagctgggg cagcaaaggc gaacatcacg ggCtgCCgCC gctataggct tgccactcct acgcaagt ct tctccaggac cctccaaccc tcaggcacct hctggtccgt ggctgagatg gtctccagag tccctgaatg tataaagtcca cttgccctgt tgcaaagccc gagcagtgct tgcagcttga tgctgtgaca atccttgcgc ctggggggcc cacacacccg gaccatgtat tcccacccgg tctctgctgc cttcccccag ggtgtCCCCC aagtggactq atggggcctg gcagcctcag cctgaggccc tgatggcagg aggtgagcaa ggaccgcgcg actgcgtgga ccgacttgtg tgctccctgc ccgctgcagc gcccagtggg gtCtgCgCCC cagatcggct tgtttccatg gaaqccttcc gcacccagca agtagaactg gaggcctgga cacagcgtag tctccaccac cttggccctg cgacjgactgc catccgcgca tgactcacag caacgccagc actcggcctg ccacactggg agcctgtcct ctgtccccca ctattgacac gcccagcatt ctgcccacc ggggacaggc gaggacagga ggaagggcc gcccttaata agcccaccag cagccaggca CtgCaggtgg gttggCCtCC gactactacg ggggcccatg ctgctctggg tgtggtgccc ggttCCtgag ccctgaccct agatccgcct ctccaccctt catctatgac act caggagg gtcgacgtqa aggcctcaca aacacctgtt tgaccatgaa ctgCCCtgCt agaactgcac tgaccgtcat tgggcaagaa ccctgcagcc gacccggcca caggcct ctg gcacatccta CCCatggCCC gcagatggcc aaccctgtgc ttgagccagg gc ccggt aaa gttcctggga ttCgtggggC ggataagcca 120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 Table Ul(a). Nucleotide sequence alignment of PSCA v.2 (SEQ ID NO:6528) and PSCA v.1 (SEQ ID NO:6529) v.2 16 agtcacctgaggccctctccaccacagcccaccagtgaccatgaaggctg v. 1 1 aggga---- cagtgaccatgaaggctg 27 v.2 66 tgctgcttgccctgttgatggcaggcttggccctgcagccaggcactgcc 115 v.1 28 tgctgcttgccctgtgatggcaggcttggccctgcaccaggcactgcc 77 v.2 116 ctgctgtgctactcctgcaaagcccagqtqagcaacgaggactgcctgca 165 v.1 78 ctgctgtgctactcctgcaaagcccaggtgagcaacgaggactgcctgca 127 v.2 166 ggtggagaactgcacccagctgggggagcagtgctggaccgcgcgcatcc 215 v.1 128 ggtggagaactgcacccagctgggggagcagtgctggaccgcgcgcatcc 177 v.2 216 gcgcagttggcctcctgaccgtcatcagcaaaggctgcagcttgaactgc 265 v.1 178 gcgcagttggcctcctgaccgtcatcagcaaaqgctgcagcttgaactgC 227 v.2 266 gtggatgactcacagcactactacgtjggcaagaagaacatcacgtgctg 315 v. 1 228 gtggatgactcacaggactactacgtgggcaagaagaacatcacgtgctg 277 v.2 316 tgacaccgacttgtgcaacgccagcggggcccatgccctgcagccggctg 365 v.1 278 tgacaccgacttgtgcaacgccagcggggcccatgccctgcagceggctg 327 v.2 366 ccgccatccttgcgctgctccctgactcggcctgctgctctggggaccc 415 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 v.1 328 ccgccatccttgcgctgctccctgcactcggcctgctgctctggggaccc 377 v.2 416 ggccagctataggctctqgggggccccgctgcacgcccacactgggtgtgg 465 v.1 378 ggccagctataggctctggggggccccgctgcagcccacactgggtgtgg 427 v.2 466 tgccccaggcctctgtgccactcctcaca-cacccggcccagtgggagcc 514 v.1 428 tgccccaggcctttgtgccactcctcacagaacctggcccagtgggagcc 477 v. 2 515 tgtcctggttcctgaggcacatcctaacgcaagtctgaccatgtatgtct 564 v.1 478 tgtcctggttcctgaggcacatactaacgcaagtttgaccatgtatgttt 527 v.2 565 gcgcccctgtccccc--accctgaccctcccat-ggccctctccaggact 611 vA 528 gcaccccttttccccnaaccctgaccttcccatgggccttttccaggatt 577 v.2 612 cccacccggcagatcggctctattgacacagatccgcctgcagatggccc 661 V. 1 578 cccacccggcaga'tcagttttagtgacacagatccgcctgcagatggccc 627 v.2 662 ctccaaccctctctgctgctgtttccatggcccagcattctccaccctta 711 v. 1 628 ctccaaccctttctgttgctgtttccatggcccaqcattttccaccctta 677 v.2 712 accctgtgctcaggcacctcttcacccaggaagccLccctgcccacccc 761 v. 1 678 accctgtgttcaggcacttcttcccecaggaagccttccctgcocacccc 727 v.2 762 atctatgacttgagccaggtctggtccgtggtgtcccccgcacccagcag 811 v. 1 728 atttatgaattgagccaggtttggtccgtggtgtcccccgcacccagcag 777 v.2 812 gggacaggcactcaggagggcccggtaaaggctgagatgaagtggactga 861 v.1 778 gggacaggcaatcaggagggcccagtaaaggctgagatqaagtggactga 827 v.2 862 gtaqaactggaggacaqgagtcgacgtgagttcctgggagtctccagaga 911 v. 1 828 gtacaactggaggacaagagttgacgtgagttcctgggagtttccagaga 877 v.2 912 tggggcctggaggcctggaggaaggggccaggctcacaBttcgtggggct 961 v.1 878 tggggcctggaggcctggaggaaggggccaggcctcacatttgtggggct 927 v. 2 962 ccctgaatggcagcctcagcacagcgtaggcccttaataaacacctgttg 1011 v.1 928 ccc-gaatggcaqcctgagcacagcgtaggcccttaataaacacctgttg 976 v.2 1012 gataagcca 1020 v.1 977 gataagcca 985 Table LIV(a). Peptide sequences of protein coded by PSCA v.2 (SEQ ID NO:6530) MKAVLLALLM AGLALQPGTA LLCYSCKAQV SNEDCLQVEN CTQLGEQCWT ARIRAVGLLT VISKGCSLNC VDDSQDYYVG KKNITCCDTD LCNASGAHAL QPAAAILALL PALGLLLWGP 120
GQL
WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Table LV(a). Amino acid sequence alignment of PSCA v.2 (SEQ ID NO:6531) and PSCA v. (SEQ ID NO:6532) v.2 1 MAVLLAILLAGLALQPGTALLCYSCKAQVSNDCLQVENCTQLGEQCWT v.1 1 MKlAVLLALLMAGLALQPGTALLCYSCKAQVSNEDCLQVENCTQLGEQCWT v.2 51 ARIRAVGLLTVISKGCSLNCVDDSQDYYVGKKNITCCDTDLCNASGAHAL 100 .1 51 ARIRAVGLLTVISKGCSLNCVDDSQDYYVGKKNITCCDTDLCNASGAHAL 100 v.2 101 QPAAAILALLPALGLLLWGPGQL V.1 101 QPAAAILALLPALGLLLWGPGQL Table 1.11(b). Nucleotide sequence of transcript variant PSCA 0. (SEQ ID NO:6533) tttgaggcca ggctgtgctg gtgctactcc agcttgaact tgtgacaccg 99ggggcccc cacacccggc ccatgtatgt ccacccggca tctgctgctg tcccccagga tgtCCCCCgC gtggactgag ggggcctgga agcctcagca tataaagtca cttgccctgt tgcaaagccc gqtgqatga acttgtgcac gCtgcagcCC ccagtgggag ctqgcccct gacggCizct tttccatggc agccttccct acccagcagg tagaactgga ggcctggagg cagcgtaggc cctgaggccc tgatgqcagg aggcgcagtt ctcacaggac tCggcctqct acactgggtg cctgtcctgg gtcccccacc attgacacag ccagcatt at gcccacccca ggacaggcac ggacaggagt aaggggccag ccttaataaa tctccaccac agcccaccag ctcggccctg ggcctq~ta tactacgtgg gCtCtgggga tggtgcccca ttcctjaggc ctgaccctcc atccgcctgc ccacccttaa tctatgac-zt t caggagggc cgacgtgagt gcct cacat cacctgttgg cagccaggca ccgt catcag gcaagaagaa cccggccagc ggcctctgtg acatcctaac catggccctc agatggcccc CCCtgtgCtC gagccaggtc ccggtaaagcj tcctgggagt CgtggggCtC at a agcca tgaccatgaa CtgCCCtgCt caaaggotgc catcacgtgc tataggctct ccactcctca gcaagtctga tccaggactc tccaaccctc aggcacctct tggtCCgtgg ctgagatgaa ctccagagat cctgaatggc Table 1.II1(b). Nucleotide sequence alignment of PSCA v.2 (SEQ ID NO:6534) and PSCA v.3 (SEQ ID NO:6535) v. 2 1 tttgaggccatataaagtcacctgaggccctctccaccacagcccaccag v.3 1 tttgaggccatataaagtcacctgaggcccttccaccacagcccaccag v.2 51 tgaccatgaaggctgtgctgcttgccctgttgatggcaggcttggccctg 100 v.3 51 tgaccatqaaggctgtgctgcttgccctgttatggcaggcttggccctg 100 v.2 101 cagccaggcactgccctgctgtgctactcctgcaaagcccaggtgagcaa 150 v. 3 101 cagccaggcactgccctgctgtgctactcctgcaaagcccag 142 v.2 151 cgaggactgcctgcaggtggagaactgcacccagctgggggagcagtgct 200 v.3 142 v.2 201 ggaccgcgcgcatccgcgcagttggcctcctgaccgtcatcagcaaaggc v. 3 gcgcagttggcctcctgaccgtcatcagcaaaggc v.2 251 tgcagcttgaactgcgtggatgactcacaggactactacgtgggcaagaa v.3 178 tgcagcttgaactcjcgtggatgactcacaggactactacgtgggcaagaa v.2 301 gaacatcacgtgctgtgacaccgacttgtgcaacgccagcgggg~ccatg WO 2005/014780 WO 205/04780PCT/1§S2004/017231 v.3 228 gaacatcacgtgctgtgacaccgacttg------------------------- 255 v.2 351 ccctgcagccqctgccgccatccttgcqctgctccctgcactcggcctg 400 v.3 tgcactcggcctg 268 v.2 401 ctgctctggggacccggccagctataggctctgggggqccccgctgcagc 450 v.3 269 ctgctctggggacccggccagctataggctctgggggqccccgctgagc 318 v.2 451 ccacactgggtgtggtgccccaggcctctgtgccactcctcacacacccg 500 v.3 319 ccacactgqgtgtggtgccccaggcctctgtgccactcctcacacacccg 368 v.2 501 gcccagtgggagcctgtcctggttcctgaggcacatcctaacgcaagtct 550 v.3 369 gcccagtgggagcctgtcctggttcctgaggcacatcctaacgcaagtct 418 v.2 551 gaccatgtatgtctgcgcccctgtcccccaccctgaccctccCatggccc 600 v.3 419 gaccatgtatgtctgcgcccctgtcccccaccctgaccctcccatggccc 468 v.2 601 tctccaqgactcccacccgqcaqatcggctctattgacacagatccgcct 650 U-3 469 tct-c-;cjggactcccacccggcagatcggctctattgacacagatccgcct 518 v.2 651 gcagatqgcccctccaaccctctctgctgctgtttccatggcccagcatt 700 v.3 519 gcagatggcccctccaaccctctctgctgctgtttccatggcccagcatt 568 v.2 701 ctccacccttaaccctgtgctcaggcacctcttcccccaggaagccttcc 750 v.3 569 ctccacccttaaccctgtgctcaggcacctcttcccccaggaagccttcc 618 v.2 751 ctgcccaccccatctatgacttgagocaggtztggtccgtggtgtccccc 800 v.3 619 ctgcccaccccatctatgacttgagccaggtctggtccgtggtgtccccc 668 v.2 801 gcacccagcaggggacaggcactcaggagggcccggtaaaggotgagatg 850 v. 3 669 gcacccagcaggggacaggcactcaggagggcccggtaaaggctgagatg 718 v.2 851 aagtggactgagtagaactggaggacaggagtcgacgtgagttcctggga 900 v.3 719 aagtggactgagtagaactgqaggacaggagtcgacgtgagttcctggga 768 v.2 901 gtctccagagatggggcctggaggcctggaggaaggggccaggctcaca 950 v.3 769 gtctccagagatggggcctggaqgcctgaggaaggqgccaggcctcaca 818 v.2 951 ttcgtggggctccctgaatggcagcctcagcacagcgtaggcccttaata 1000 v.3 819 ttcgtggggctccatgaatggcagcctcagcacagcgtaggcccttaata 868 v.2 1001 aacacctgttggataagcca 1020 v.3 869 aacacctgttggataagcca 888 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 Table LIV(b). Peptide sequences of protein coded by PSCA v.3 (SEQ ID NO:6536) MYVCAPVPHP DPBL4ALSRTP TRQIGSIDTD PPADGPSNPL CCCFHGPAFS PQEAFPAHPI YDLSQVWSVV SPAPSRGQAL RRAR Table LV(b). Amino acid sequence alignment of PSCA v.2 and PSCA v.3 NO SIGNIFICANT HOMOL~OGY Table 1.11(c). Nucleotide sequence of transcript variant PSCA v.4 (SEQ ID NO:6537)
TLNPVLRHLY
gacagtgaac cccagggttt agggcagtgc acttagctgg tttgaactgg gcaacgagga cgcgcatccg tggatgactc tgtgcaacgc ctgcactcgg cagc ocac ac tgggagcctg gCCCCtgtCC ggctctattg catggcccag ttCCCtgCCC agcaggggac actggaggac tqgaggaagg gtaggccctt cctgcgctga cgtgccgatc tgccttccgg ggtcaaatcc gtgcgactta ctgcctgcag cgcagttggc acaggactac cagCggggCC cctgctgctc tgggtgtggt tcctggttcc cccaccctga acacagatcc cattctccac accccatcta aggoactcag aggagtcgac ggccaggcct aataaacacc aggcgttgg agcccaggac tcaccaggac atacccaatt agcactgccc gtggagaact ctcctgaccg tacgtgggca catgccctgc tggggacccg gccccaggcc tgaggcacat ccctcccatg gcctgcagat ccttaaccct tgacttgagc gagggcccg gtgagttcct cacattcgtg tgttggataa gctcctgcaq ggtcttcccg cagtgctcag tagatgatc tgctgtgcta gcacccagct tcatcagcaa agaagaacat agccggctgc gccagctata tctqtgccac cctaacgcaa gccctctcca ggCCCCtCCa gtgct CaggC caggtctggt taaaggctga gggagtctcc gggCtccctg gcc a ttctggggca gtgcagtttc cccqcctgct agacgatggg ctcctgcaaa gggggagcag aggctgcagc cacgtgctgt cgCCatCCtt ggCtctgggg tcctcacaca gtctgaccat qqactcccac accctctctg acctcttccc ccgtggtgtc gatgaagtgg agagatgggg aatggcagcc gccacaggcg tgatgcgggg tgaccccctt atttgaaact gcccaggtga tgctggaccg ttgaactgcg.
gacaccgact gCgCtgCtCC ggccccgctg cccggcccag gtatgtctgc ccggcagat c ctgctgtttc ccaggaagcc ccccgcaccc actgagtaga cctggaggcc tcagcacagc 120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1174 Table 1-111(c). Nucleotide sequence alignment of PSCA v.2 (SEQ ID NO:6538) and PSCA v.4 (SEQ ID NO:6539) v. 2 1 tttgaggccatataaagtcacctgagqccctctccacca 39 v. 4 42 tctggggc agocac aggcgc ccagggtttcgtgc v.2 40 cagccca ccagtgacca tgaag 61 v. 4 76 cgatcagcccaggacggtcttcccggtg--cagtttctgatgcggggagg 123 v. 2 62 gctgtgctg-cttgccctgt tgatggcag gc 91 v. 4 124 gcagtgctgcctt--ccggtcaccaggaccagtgct--cagcccgcctgc 169 v. 2 92 tg 100 v.4 170 ttgacccccttacttagctggggtcaatccatac-ccaattbagatgatt 219 v. 2 101 aggcactgcc 115 v. 4 220 cagacgatgggatttgaaacttttgaactgggtgcgacttaagcactgcc 269 v.2 116 ctgctgtgctactcctgcaaagcccaggtgagcaacgaggactgcctgca 165 v.4 270 ctgctgtgctactcctgcaaagcccaggtgagcaacgaggactgcctgca 319 v.2 166 ggtggagaactgcacccagctgggggagcagtgctggaccgcgcgcatcc 215 v.4 320 ggtggagaactgcacccagctgggggagcagtgctggaccgcgcgcatcc 369 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 v.2 216 gcgcagttggcctcctgaccgtcatcagcaaaggctgcagcttgaactgc 265 v.4 370 gcgcagttggcctcctgaccgtcatcagcaaaggctgcagcttgaactgc 419 v.2 266 gtggatgactcacaggactactacgtgggcaagaagaacatcacgtgctg 315 v.4 420 gtggatgactcacaggactactacgtgggcaagaagaacatcacgtgctg 469 v.2 316 tgacaccgacttgtgcaacgccagcggggcccatgccctgcagccggctg 365 v.4 470 tqacaccqacttgtgcaacqccagcggggccaatqccctgcagccggctg 519 v.2 366 ccgccatccttgcgctgctccctgcactcggcctgctgctctggggaccc 415 v.4 520 ccgccatccttgcgctgctccctgcactcggcctgctgctctgqgqaccc 569 v.2 416 ggccagctataggctctgqgggccccgctgcagcccacactgggtgtgg 465 v. 4 570 ggccagctataggctctggqgggccccgctgcagcccacactgggtgtgg 619 v.2 466 tgccccaggcctctgtgccactcctcacacacccggcccagtgggagcct 515 v.4 620 tgccccaggcctctgtgccactcctcacacacccggcccagtgggagcct 669 v.2 516 gtcctggttcctgaggcacatcctaacgcaagtctgaccatgtatgtctg 565 v.4 670 gtcctggttcctgaggcacatcctaacgcaagtctgaccatgtatgtctg 719 v.2 566 cgcccctgtcccccaccctgaccctcccatggccctctccaggactccca 615 VA4 720 cgcccctgtcccccaccctgaccctcccatggccctctccaggactccca 769 v.2 616 cccggcagatcggctctattgacacagatccgcctqcagatggccctcc 665 v.4 770 cccggcagatcggctctattgacacagatccgcctgcagatggcccctcc 819 v.2 666 aaccctctctgctgctgtttccatggcccagcattctccacccttaaccc 715 v.4 820 aaccctctctgctgctgtttccatggcccagcattctccacccttaacCC 869 v.2 716 tgtgctcaggcacctcttcccccaggaagccttccctgcccaccccatct 765 v.4 870 tgtgctcaggcacctcttcccccagcjaagccttccctgcccaccccatct 919 v.2 766 atgacttgagccaggtctggtccgtggtgtecccccgcacccagcagggga 815 v.4 920 atgacttgagccaggtctggtccgtggtgtccacccgcacccagcagggga 969 v.2 816 caggcactcaggagggcccggtaaaggctgagatgaagtggjactgagtag 865 v.4 970 caggcactcaggagggcccggtaaaggctgagatgaagtggactgagtag 1019 v.2 866 aactggaggacaggaqtcgacgtgagttcctgggagtctccagagatggg 915 v.4 1020 aactggaggacaggagtcgacgtgagttcctgggagtctccagagatggg 1069 v.2 916 gcctggaggcctggaggaaggggccaggcctcacattcgtggggctcccL 965 236 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 v.4 1070 gcctggaggcctggaggaaggggccaggcctcacattcgtggggctccct 1119 v.2 966 gaatggcagcctcagcacagcgtaggcccttaataaacacctgttggata v.4 1120 gaatggcagcctcagcacagcgtaggcccttaataaacacctgttggata 1015 1169 v.2 1016 agcca 1020 v.4 1170 agcca 1174 Table LIV(c). Peptide sequences of protein coded by PSCA v.4 (SEQ ID NO:6540) MTHRTTTWAR RTSRAVTPTC ATPAGPMPCS RLPPSLRCSL HSACCSGDPA SYRLWGAPLQ PTLGVVPQAS VPLLTHPAQW EPVLVPEAHP NASLTMYVCA PVPHPDPPMA LSRTPT-RQIG SIDTDPPADG PSNPLCCCFH GPAFSTLNPV LRHLFPQEAF PAHPIYDLSQ VWSVVSPAPS RGQAkLRPAR Table LV(c). Amino acid sequence alignment of PSCA v.2 and PSCA v.4 NO SIGNIFICANT HOMOLOGY Table 1-lI(d). Nucleotide sequence of transcript variant PSCA v.5 (SEQ ID NO:6541) gacagtgaac cccaggqttt agggcagtgc acttagctgg tttgaactg gcaacgagga cgcgcatccg taatctttct cgtcccgcacatctgccact tctgtccctc gtggtccatc gcaCtgCttg caggaggagg cccgctgctc actgcgtgcja ccgacttgtg Lgctccctgc ccgctgcagc gcccagtggg gtctgcgccc cagatcggct tgtttCCatg gaagccttcc gcacccagca agtagaactg gaggcctgga cacagcgtag cctgcgctga cgtgccgatc tgccttccgg ggtccaatcc gtgcgactta ctgcctgca9 tgagtggggg ggccatctgt ctgcaccccc cctccactca cccatccctg tgcctcggac caatcctgag cctggggcag cccaggcgca tgactcacag caacgccagc actcggcctg ccacactggg agCCtgtCCt ctgtccccca ctattgacac gcccagcatt ctgcccaccc ggggacaggc gaggacagga ggaaggggcc qcccttaata aggcgttgg agcccaggac tcaccaggac atacccaatt agcactgccc gtggagaact gacgacagcc ccgcatctgt aacaatcacc tctgtccctc agctcactta at ctggat ag geccagc cc ag gtcaggcaq 9ttggcctcc gactactacg ggggcccatg ctgctctggg tgtggtgccc ggttCCtgag ccctgaccct agatccgcct ot ccaccctt catctatgac actcaggagg gicgacgtga aggcctcaca aacacctgtt gctcctgcag ggtcttcccg cagt gctcag tagatgattc tgctgtgcta gcacccagct gcoaggccc-a gtgctgttzt cagcatctgt cccatcctcc ctcactcacc ggctgagacc ggggactcta tgagcacaca tgaccgtcat tgggcaagaa ccctgcagcc gacccggcca cagqcctctg gcacatccta cccatggccc gCagatgg~C aaccctgtgc ttgagccagg gcccggtaaa gttcctggga ttcgtggggc ggataagcca ttctggggca gtgcagtttc cccgcctgct aqacqatg ctcctgcaaa gggggagcag ggtctctgcc ccttccacct ccctccagcc atcttccact ccat ttctga agggccgaga gagcattagg gggcagcccc cagcaaaggc gaacatcacq ggctgccgcc gctataggct tgccactcct acgcaagtct tctccaggac cctccaaccc tcaggcacct tctggtccgt ggctgaqatg gtctccagag tccctgaatg gccacaggcg tgatgcgggg tcjaccccctt atttgaaact gcccaggtga tgCtggaCCg actgaactat gtccccgacc atcetccatcc cctccaccca cgctcagcgg ccaggccctc cagggtggga atccccggat tgcagcttga tgctgtgaca atccttgcgc Ctggggggcc cacacacccg gaccatgtat t cccacccgg tctctgctgc cttcccccag ggtgt ccccc aagtggactg atggggcctg gcagcctcag 120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1660 Table 1.I1I(d). Nucleotide sequence alignment of PSCAv.2 (SEQ ID NO:6542 and PSCAv.5 (SEQ ID NO:6543) v.2 1 tttgaggccatataaagtcacctgaggccctctccacca 39 v. 5 42 tctggggc agccac aggcgc ccaggtttcgtgc v. 2 40 cagccca ccagtgacca tgaag 61 111111 11-111 11 .11.1 76 cgatcagcccaggacggtcttcccggtg--cagtttctgatgcggggagg 123 WO 2005/014780 PCT/1S2004/017231 v.2 62 gctgtgctg-cttgccctgt-------------tgatggcag-------gc 91 III111 .1 11.1 I III II 124 gcagtgctgcctt--ccggtcaccaggaccagtgct--cagcccgcctgc 169 v.2 92 100 I II. I I.
170 ttgacccccttacttagctggggtccaatccatacccaatttaqatqatt 219 v.2 101 115 220 cagacgatgggatttgaaacttttqaactgggtgcgacttaagcactgcc 269 v.2 116 ctgctgtgctactcctgcaaagcccaggtgagcaacgaggactgcctgca 165 I 1III I 11111111 I /1111111111111 III 1111111 270 ctgctgtgctactcctqcaaagcccaggtgagcaacgaggactgcctgca 319 v.2 166 ggtggagaactgcacccagctgggggagcagtgctggaccgcgcatcc 215 11111111 111111111 li 1111111111111 1111111 IIIlll 320 ggtggagaactgcacccagctgggggagcagtgctggaccgcgcgcatCC 369 v.2 216 215 370 gtgagtggggggacgacagccgccaggcctaggtctctgccactgaacta 419 v.2 216 215 420 ttaatctttctggccatctgtccgcatctgtgtgctgttttccttceacc 469 v.2 216 215 470 tgtccccgacccgtcccgcacctgcacccccaacaatcaccoagcatctg 519 v.2 216 215 520 tccctccagccatcctcctccatctgccactcctccactcatctgtccct 569 v.2 216 215 570 ccccatcctccatcttccactcctccacccatctgtcctcCcc atccct 619 v.2 216 215 620 gagctcacttactcactcaccccatttctgacgctcagcgggtggtccat 669 v.2 216 215 670 ctgcctcggacatctggatagggctgagaccagggccgagaccagccct 719 v.2 216 215 720 cgcactgcttgcaatcctgaggccagcccagggggactctagagcattag 769 v.2 216 215 770 gcagggtgggacaggaggaggcctggggcaggtcaggcaggtgagcaca 819 v.2 216 gcgcagttggcctc 229 WO 2005/014780 WO 205/04780PCT/1§S2004/017231 820 agggcagccccatccccqgatcccgctgctccccaggcgcagttggcctc 86 v.2 230 ctgaccgtcatcagcaaaggctgcagcttgaactgcgtggatgactcaca 279 870 ctgaccgtcatcagcaaaggctgcagcttgaactgcgtggatgactcaca 919 v.2 280 ggactactacgtgggcaagaagaacatcacgtgctgtgacaccgacttgt 329 920 ggactactacgtgggcaagaagaacatcacgtgctgtgacaccgacttgt 969 v.2 330 gcaacgccagcggggcccatgccctgcagccggctgccqccatCCttgcg 379 970 gcaacgacagcggggcccatgccctgCagccggctgccgccatccttgcg 1019 v.2 380 ctgctccctgcactcggcctgctgctctggggacccggccagctataggc 429 1020 ctgctccctgcactcggcctgctgctctggggacccggccagctataggc 1069 v.2 430 tctggggggccccgctgcagcccacactgggtgtggtgccccaggcctct 479 1070 tctggggggccccgctgcagcccacactgggtgtggccccagqcctct 1119 v.2 480 gtgccactcctcacacacccggcccagtgggagcctgtcctggttcctga 529 1120 gtgccactcctcacacacccqgcccatgggagcctgtcctggttcctga 1169 v.2 530 ggcacatcctaacgaaagtetgaccatgtatgtctgcgcccctgtccccc 579 1170 ggcacatcctaacgcaagtctgaccatgtatgtctqgcccctgtcCccc 1219 v.2 580 accctgaccctcccatggccctctccaqgactcccacccggcagatcggc 629 1220 accctgaccctcccatggccctctccaggactcccacccggcagatcggc 1269 v.2 630 tctattgacacagatcgcctgcagatggcccctcaaccctctctgctg 679 1270 tctattgacacagatccgcctgcagatggcccctccaaccctctctgctg 1319 v.2 680 ctgtttccatggcccagcattctccacacttaaccctgtgctcaggcacc 729 1320 ctgtttccatggcccagcattctccacccttaaccctgtgctcaggcacc 1369 v.2 730 tcttcccccaggaagccttccctgcccaccccatctatgacttgagCCag 779 1370 tcttccccaggaagccttccctgcccaccccatctagacttagCCag 1419 v.2 780 gtctggtccgtggtgtcccccgcacccagcaggggacaggcactcaggag 829 1420 gtctggtccgtggtgtcccccgcacccagcaggggacaggcactcaggag 1469 v.2 830 ggcccggtaaaggctgagatgaagtggactgagtagaactggagacagg 879 1470 ggcccggtaaaggctgagatgaagtggactgagtagaactgaggacagg 1519 v.2 880 agtegacgtgagttcctggagtctccagagatggggcctggaggcctgg 929 v. 5 1520 agtcgacgtgagttcctgggagtctccagagatggggCtgaggcctgg 1569 v.2 930 aggaagqggccaggcctcacattcgtggggct~ccctgaatggcagcctca 979 239 WO 2005/014780 PCT/1S2004/017231 1570 aggaaggqgccaii 111111111111111cccc IIIaggca 1570 aggaaqgggccaggcctcacattcgtggggctccctgaatgcgta 1619 v.2 980 gcacagcgtaggcccttaataaacacctgttggataagcca v.51 1601111111ggcctataa I 1111111 I IIII 1620 gcacagcgtaggcccttaataaacacctgttggataagcca 1020 1660 Table LIV(d). Peptide sequences of protein coded by PSCA v.5 (SEQ ID NO:6544) MTHRTTTWAR RTSRAVTPTC ATPAGFMPCS RLPPSLRCSL HSACCSGDPA SYRLWGAPLQ PTLSVVPQAS VPLLTPAQW EPVLVPEAIP NASLTMYVCA PVPHPDPPMA LSRTPTRQIG SIDTDPPADG PSNPLCCCFH GPAFSTLNPV LRHLFPQEAF PAHPIYDLSQ VWSVVSPAPS
RGQALRRAR
Table LV(d). Amino acid sequence alignment of PSCA v.2 and PSCA NO SIGNIFICANT HOMOLOGY Table LVI SNP and codon changes in PSCA v.2 and v.4 Variant V.2 SNP AA* AA V.4 AA AA Variant position change position position change position V.6 57 t/c 1 Not in v.4 V.7 367 c/t A/A 104 521 P/L 33 v.19 V.8 424 a/c UL 123 578 Y/S 52 V.9 495 clg 649 H/f 76 v.21 499 c/t 653 P/L 77 v.22 V.11 563 c/t 717 VIV 98 v.23 V.12 567 gla 721 AI 100 v.24 V.13 627 g/a 781 G/S 120 V.14 634 t/g 788 I/S 122 v.26 835 gla 989 R/Q 189 v.27 V.16 847 g/a 1001 v.28 V.17 878 g/a 1032 v.29 V.18 978 c/g 1132 AA: amino acid No amino acid encoded.

Claims (44)

1. 1. An isolated polynucleotide that encodes a PSCA protein, wherein the polynucleotide is selected from the group consisting of: a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 521 is T; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 578 is C; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 649 is G; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 653 is T; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 721 is A; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 781 is A; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 788 is G; a polynucleotide comprising the sequence of SEQ ID NO:6537, from nucleotide residue numbers 424 through 993, wherein the nucleotide residue at 989 is A; a polynucleotide comprising the sequence of SEQ ID NO:6533, from nucleotide residue numbers 423 through 707; and a polynucleotide comprising the sequence of SEQ ID NO:6545, from nucleotide residue numbers 83 through 427. 00 O O
2. A polynucleotide that is fully complementary to a polynucleotide of any one of &Jb of claim 1.
3. A polynucleotide of claim 1 that encodes the polypeptide sequence shown in SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546. 00
4. A polynucleotide of claim 1 that encodes the polypeptide sequence shown in M SEQ ID NO:6540 wherein P33L, Y52S, H76D, P77L, A100T, G120S, I122S, or N R189Q.
5. A recombinant expression vector comprising a polynucleotide of any one of claims 1-4.
6. The expression vector of claim 5, wherein said vector is a viral expression vector.
7. The expression vector of claim 6, wherein the viral expression vector is derived from a vaccinia virus, fowlpox virus, canarypox virus, adenovirus, influenza virus, poliovirus, adeno-associated virus, lentivirus, or sindbis virus
8. A host cell that contains an expression vector of any one of claims 5 to 7.
9. A process for producing a PSCA protein comprising culturing a host cell of claim 6 under conditions sufficient for the production of the PSCA protein.
The process of claim 9, further comprising recovering the PSCA protein so produced.
11. The process of claim 10, wherein the protein is recovered using chromatography.
12. The process of claim 10, wherein the protein is recovered using metal chelate chromatography.
13. An isolated PSCA protein, wherein the PSCA protein comprises SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546. 00 O O
14. A composition comprising a pharmaceutically acceptable carrier and a protein Sof claim 13. C
15. An antibody or fragment thereof that immunospecifically binds to an epitope on a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, c 6536, or 6546). 00 M
16. The antibody or fragment thereof of claim 15, which is monoclonal. O 10
17. A hybridoma that produces an antibody of claim 16.
18. The antibody or fragment thereof of claim 16, wherein the monoclonal antibody is a recombinant protein.
19. The antibody or fragment thereof of claim 18, which is a single chain monoclonal antibody.
The antibody or fragment thereof of claim 15, which is a human antibody.
21. The antibody or fragment thereof of any one of claims 15, 16, 17, 19, or which is labeled with an agent.
22. The antibody or fragment thereof of claim 21, wherein the cytotoxic agent is selected from the group consisting of radioactive isotopes, chemotherapeutic agents and toxins.
23. The antibody or fragment thereof of claim 22, wherein the radioactive isotope is selected from the group consisting of 2 11 At, 131I, 125, 90 Y, 1 86 Re, 88 Re, 53Sm, 2 12 Bi, 32 P and radioactive isotopes of Lu.
24. The antibody or fragment thereof of claim 22, wherein the chemotherapeutic agent is selected from the group consisting of taxol, actinomycin, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, gelonin, and calicheamicin.
25. The antibody or fragment thereof of claim 22, wherein the toxin is selected from the group consisting of diphtheria toxin, enomycin, phenomycin, Pseudomonas 243 00 O exotoxin (PE) A, PE40, abrin, abrin A chain, mitogellin, modeccin A chain, and alpha- tb Ssarcin.
26. The antibody or fragment thereof of any one of claims 15, 16, and 18 to wherein the antibody or fragment thereof further comprises a pharmaceutically acceptable carrier. 00 M
27. An in vitro method for detecting the presence of a PSCA protein (SEQ ID N NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546) or O 10 polynucleotide (SEQ ID NO: 6533, SEQ ID NO:6545, or SEQ ID NO:6537, wherein 0 C521T, A578C, C649G, C653T, G721A, G781A, T788G, or G989A) in a test sample comprising: contacting the sample with an antibody or polynucleotide, respectively, that specifically binds to the PSCA protein or polynucleotide, respectively; and detecting binding of PSCA protein or polynucleotide, respectively, in the sample thereto.
28. The method of claim 27, wherein the polynucleotide is an mRNA.
29. The method of claim 27, wherein the polynucleotide is a cDNA produced from the sample by reverse transcription.
The method of any one of claims 27 to 29, wherein the determining step comprises comparing an amount of binding of the antibody or polynucleotide that specifically binds to the PSCA protein or polynucleotide to the presence of the protein or polynucleotide in a corresponding normal sample.
31. The method of claim 30, wherein the presence of elevated PSCA polynucleotide or protein in the test sample relative to the normal tissue sample provides an indication of the presence of cancer.
32. The method of claim 31, wherein the cancer is selected from the group consisting of leukemia and cancer of the prostate, testis, kidney, brain, bone, skin, ovary, breast, pancreas, colon, and lung, and the test and normal tissue samples are selected from the group consisting of serum, blood or urine and tissues of the prostate, testis, kidney, brain, bone, skin, ovary, breast, pancreas, colon, and lung. 00 O
33. An in vitro method of inhibiting growth of a cell expressing a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546), comprising providing an effective amount of an antibody according to any one of Sclaims 15, 16, or 18 to 25 to the cell, whereby the growth of the cell is inhibited. Cc
34. An in vitro method of delivering a cytotoxic agent to a cell expressing a PSCA 00 protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or M 6546) comprising providing an effective amount of an antibody according to any one of N claims 20 to 24 to the cell. O C
35. A method of inducing an immune response to a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546), said method comprising: providing a PSCA protein epitope; contacting the epitope with an immune system T cell or B cell, whereby the immune system T cell or B cell is induced.
36. The method of claim 35, wherein the immune system cell is a B cell, whereby the induced B cell generates antibodies that specifically bind to the PSCA protein.
37. The method of claim 35, wherein the immune system cell is a T cell that is a cytotoxic T cell (CTL), whereby the activated CTL kills an autologous cell that expresses the PSCA protein.
38. The method of claim 35, wherein the immune system cell is a T cell that is a helper T cell (HTL), whereby the activated HTL secretes cytokines that facilitate the cytotoxic activity of a CTL or the antibody producing activity of a B cell.
39. Use of a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546) epitope for the preparation of a medicament to induce an immune response in a subject.
The use of claim 39, wherein the immune response comprises activation of a B cell, wherein the activated B cells generate antibodies that specifically bind to the PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546). 00 O O
41. The use of claim 39, wherein the immune response comprises activation ofa T Scell, wherein the activated T cell is a cytotoxic T cell (CTL), which, when activated kills an autologous cell that expresses the PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546).
42. The use of claim 39, wherein the immune response comprises activation of a T 00 cell, wherein the activated T cell is a helper T cell (HTL), which, when activated M secretes cytokines that facilitate cytotoxic activity of a CTL or antibody producing activity ofa B cell. O C
43. Use of an antibody for the preparation of a medicament which delivers an agent to a cell expressing a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546), wherein the antibody comprises an antibody according to any one of claims 21 to 26.
44. Use of an effective amount of an antibody according to any one of claims 16, or 18 to 26 for the preparation of a medicament which inhibits growth of a cell expressing a PSCA protein (SEQ ID NO:6540, 6547, 6548, 6549, 6550, 6551, 6552, 6553, 6554, 6536, or 6546).
AU2004263823A 2003-05-30 2004-05-28 Prostate stem cell antigen (PSCA) variants and subsequences thereof Expired AU2004263823B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2008258185A AU2008258185B2 (en) 2003-05-30 2008-12-17 Prostate stem cell antigen (PSCA) variants and subsequences thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US47506403P 2003-05-30 2003-05-30
US60/475,064 2003-05-30
PCT/US2004/017231 WO2005014780A2 (en) 2003-05-30 2004-05-28 Prostate stem cell antigen (psca) variants and subsequences thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2008258185A Division AU2008258185B2 (en) 2003-05-30 2008-12-17 Prostate stem cell antigen (PSCA) variants and subsequences thereof

Publications (2)

Publication Number Publication Date
AU2004263823A1 AU2004263823A1 (en) 2005-02-17
AU2004263823B2 true AU2004263823B2 (en) 2008-09-18

Family

ID=34135042

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2004263823A Expired AU2004263823B2 (en) 2003-05-30 2004-05-28 Prostate stem cell antigen (PSCA) variants and subsequences thereof
AU2008258185A Expired AU2008258185B2 (en) 2003-05-30 2008-12-17 Prostate stem cell antigen (PSCA) variants and subsequences thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2008258185A Expired AU2008258185B2 (en) 2003-05-30 2008-12-17 Prostate stem cell antigen (PSCA) variants and subsequences thereof

Country Status (24)

Country Link
US (4) US7622564B2 (en)
EP (2) EP2319524B1 (en)
JP (1) JP2007525196A (en)
KR (1) KR101291787B1 (en)
CN (1) CN101090729B (en)
AT (1) ATE541052T1 (en)
AU (2) AU2004263823B2 (en)
BR (1) BRPI0410842B8 (en)
CA (1) CA2526274C (en)
CY (1) CY1113044T1 (en)
DK (2) DK2319524T3 (en)
ES (1) ES2384622T3 (en)
HK (1) HK1088635A1 (en)
IL (1) IL171966A (en)
IS (1) IS2900B (en)
MX (1) MXPA05012957A (en)
NO (1) NO20055508L (en)
NZ (3) NZ578591A (en)
PL (1) PL1629088T3 (en)
PT (1) PT1629088E (en)
RU (1) RU2005141341A (en)
SI (1) SI1629088T1 (en)
WO (1) WO2005014780A2 (en)
ZA (1) ZA200509606B (en)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6541212B2 (en) * 1997-03-10 2003-04-01 The Regents Of The University Of California Methods for detecting prostate stem cell antigen protein
SI1629088T1 (en) * 2003-05-30 2012-08-31 Agensys Inc Prostate stem cell antigen (psca) variants and subsequences thereof
US7541442B2 (en) * 2003-05-30 2009-06-02 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
US7595379B2 (en) * 2003-05-30 2009-09-29 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
EP1850860A2 (en) * 2005-01-13 2007-11-07 The Johns Hopkins University Prostate stem cell antigen vaccines and uses thereof
CN101223191B (en) * 2005-04-14 2012-09-05 阿根西斯公司 Antibodies and related molecules that bind to PSCA proteins
CA2605931A1 (en) * 2005-04-26 2006-11-02 Karyon-Ctt Ltd Diagnostic and therapeutic agents
EP1973576B1 (en) * 2005-11-28 2019-05-15 Genmab A/S Recombinant monovalent antibodies and methods for production thereof
WO2007086932A2 (en) * 2006-01-13 2007-08-02 The Johns Hopkins University Prostate stem cell antigen vaccines and uses thereof
CA2646329C (en) 2006-03-20 2018-07-03 The Regents Of The University Of California Engineered anti-prostate stem cell antigen (psca) antibodies for cancer targeting
US8594203B2 (en) * 2006-10-16 2013-11-26 Nokia Corporation System and method for using parallelly decodable slices for multi-view video coding
EP2197491A4 (en) 2007-09-04 2011-01-12 Univ California High affinity anti-prostate stem cell antigen (psca) antibodies for cancer targeting and detection
EP2215122A4 (en) 2007-10-25 2011-03-09 Viventia Biotech Inc Antibodies against a cancer-associated epitope of variant hnrnpg and uses thereof
US20100069616A1 (en) * 2008-08-06 2010-03-18 The Regents Of The University Of California Engineered antibody-nanoparticle conjugates
LT2356462T (en) * 2008-11-11 2017-03-10 The Regents Of The University Of Michigan Anti-cxcr1 compositions and methods
JP5906090B2 (en) 2009-02-17 2016-04-20 コーネル・リサーチ・ファンデーション・インコーポレイテッドCornell Research Foundation, Incorporated Methods and kits for cancer diagnosis and estimation of therapeutic value
CA2782333C (en) 2009-12-02 2019-06-04 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
KR101335203B1 (en) * 2010-03-26 2013-11-29 숙명여자대학교산학협력단 Peptides for Promotion of Angiogenesis and the use thereof
US8449512B2 (en) 2010-04-09 2013-05-28 Davinci Biomedical Research Products Inc. Stoma stabilitating device and method
AU2015261742B2 (en) * 2010-11-12 2017-10-26 Inovio Pharmaceuticals, Inc. Consensus prostate antigens nucleic acid molecule encoding the same and vaccine and uses comprising the same
US8927692B2 (en) 2010-11-12 2015-01-06 The Trustees Of The University Of Pennsylvania Consensus prostate antigens, nucleic acid molecule encoding the same and vaccine and uses comprising the same
US8928126B2 (en) * 2012-11-07 2015-01-06 United Microelectronics Corp. Epitaxial layer
KR102196884B1 (en) 2013-11-01 2020-12-30 화이자 인코포레이티드 Vectors for expression of prostate-associated antigens
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
IL284985B2 (en) 2015-02-18 2023-03-01 Enlivex Therapeutics R& D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
EP3285877B1 (en) 2015-04-21 2022-10-19 Enlivex Therapeutics Rdo Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
EP4137158A1 (en) 2015-08-07 2023-02-22 Imaginab, Inc. Antigen binding constructs to target molecules
CA3014885A1 (en) 2016-02-18 2017-08-24 Enlivex Therapeutics Ltd. Combination immune therapy and cytokine control therapy for cancer treatment
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
CN106749674A (en) * 2016-12-23 2017-05-31 四川大学 A kind of new asthma polypeptide vaccine and preparation method thereof
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies
CN111951887B (en) * 2020-07-27 2024-06-28 深圳市新合生物医疗科技有限公司 Leucocyte antigen and polypeptide binding affinity prediction method based on deep learning
CN112307622B (en) * 2020-10-30 2024-05-17 中国兵器科学研究院 Autonomous planning system and planning method for generating force by computer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6258939B1 (en) * 1997-03-10 2001-07-10 The Regents Of The University Of California PSCA antibodies and hybridomas producing them
US6635468B2 (en) * 1997-09-17 2003-10-21 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same

Family Cites Families (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US337A (en) 1837-07-31 Mode of constructing railroad-frogs
US5506A (en) 1848-04-11 Jackson stjtton
US28883A (en) 1860-06-26 James
US5736A (en) 1848-08-29 Machinery fob dressing axes
US142A (en) 1837-03-11 Cooking apparatus
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
JPS6023084B2 (en) 1979-07-11 1985-06-05 味の素株式会社 blood substitute
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4722848A (en) 1982-12-08 1988-02-02 Health Research, Incorporated Method for immunizing animals with synthetically modified vaccinia virus
US4870009A (en) 1982-11-22 1989-09-26 The Salk Institute For Biological Studies Method of obtaining gene product through the generation of transgenic animals
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
DE3675588D1 (en) 1985-06-19 1990-12-20 Ajinomoto Kk HAEMOGLOBIN TIED TO A POLY (ALKENYLENE OXIDE).
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5124246A (en) 1987-10-15 1992-06-23 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using same
US5359100A (en) 1987-10-15 1994-10-25 Chiron Corporation Bifunctional blocked phosphoramidites useful in making nucleic acid mutimers
GB8724885D0 (en) 1987-10-23 1987-11-25 Binns M M Fowlpox virus promotors
US5254678A (en) 1987-12-15 1993-10-19 Gene Shears Pty. Limited Ribozymes
US5010175A (en) 1988-05-02 1991-04-23 The Regents Of The University Of California General method for producing and selecting peptides with specific properties
CA1340323C (en) 1988-09-20 1999-01-19 Arnold E. Hampel Rna catalyst for cleaving specific rna sequences
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
JPH04503957A (en) 1989-03-07 1992-07-16 ジェネンテク,インコーポレイテッド Covalent conjugates of lipids and oligonucleotides
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
WO1991004753A1 (en) 1989-10-02 1991-04-18 Cetus Corporation Conjugates of antisense oligonucleotides and therapeutic uses thereof
ATE157012T1 (en) 1989-11-03 1997-09-15 Univ Vanderbilt METHOD FOR THE IN VIVO ADMINISTRATION OF FUNCTIONAL FOREIGN GENES
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
DE69133566T2 (en) 1990-01-12 2007-12-06 Amgen Fremont Inc. Formation of xenogenic antibodies
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5204253A (en) 1990-05-29 1993-04-20 E. I. Du Pont De Nemours And Company Method and apparatus for introducing biological substances into living cells
IE66205B1 (en) 1990-06-14 1995-12-13 Paul A Bartlett Polypeptide analogs
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
ES2264239T3 (en) 1990-06-27 2006-12-16 Princeton University PROTEIN COMPLEX P53 / P90 '.
US5650489A (en) 1990-07-02 1997-07-22 The Arizona Board Of Regents Random bio-oligomer library, a method of synthesis thereof, and a method of use thereof
SE9002480D0 (en) 1990-07-23 1990-07-23 Hans Lilja ASSAY OF FREE AND COMPLEXED PROSTATE-SPECIFIC ANTIGEN
US5733731A (en) 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5541061A (en) 1992-04-29 1996-07-30 Affymax Technologies N.V. Methods for screening factorial chemical libraries
AU4528493A (en) 1992-06-04 1994-01-04 Regents Of The University Of California, The In vivo gene therapy with intron-free sequence of interest
NZ255683A (en) 1992-08-07 1996-08-27 Cytel Corp Immunogenic composition comprising a 9-10 aa residue peptide having a hla-a3.2, hla-a1, hla-a11 or hla-a24.1 binding motif
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
DE69314148T2 (en) 1992-12-17 1998-01-15 Valeo Systemes Essuyage Power supply method and device for an electric motor for driving a windshield wiper of a motor vehicle
US5576220A (en) 1993-02-19 1996-11-19 Arris Pharmaceutical Corporation Thin film HPMP matrix systems and methods for constructing and displaying ligands
NZ263050A (en) 1993-03-05 1997-11-24 Cytel Corp Compositions of immunogenic peptides with hla-a2.1 binding motifs
KR960702518A (en) 1993-05-17 1996-04-27 린다 에스, 스티븐슨 RIBOZYME GENE THERAPY FOR HIV INFECTION AND AIDS
US5837501A (en) 1993-07-09 1998-11-17 Akzo Nobel N.V. Nucleic acid quantitation by co-amplification of target with multiple internal controls
US5804566A (en) 1993-08-26 1998-09-08 The Regents Of The University Of California Methods and devices for immunizing a host through administration of naked polynucleotides with encode allergenic peptides
US5679647A (en) 1993-08-26 1997-10-21 The Regents Of The University Of California Methods and devices for immunizing a host against tumor-associated antigens through administration of naked polynucleotides which encode tumor-associated antigenic peptides
WO1995007707A1 (en) 1993-09-14 1995-03-23 Cytel Corporation Alteration of immune response using pan dr-binding peptides
US5681697A (en) 1993-12-08 1997-10-28 Chiron Corporation Solution phase nucleic acid sandwich assays having reduced background noise and kits therefor
CH686982A5 (en) 1993-12-16 1996-08-15 Maurice Stroun Method for diagnosis of cancers.
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5593853A (en) 1994-02-09 1997-01-14 Martek Corporation Generation and screening of synthetic drug libraries
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5739118A (en) 1994-04-01 1998-04-14 Apollon, Inc. Compositions and methods for delivery of genetic material
US5525735A (en) 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
EP1405911A1 (en) 1994-07-20 2004-04-07 The General Hospital Corporation Interaction trap systems for detecting protein interactions
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5681702A (en) 1994-08-30 1997-10-28 Chiron Corporation Reduction of nonspecific hybridization by using novel base-pairing schemes
WO2001004311A1 (en) * 1999-07-07 2001-01-18 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US5919652A (en) 1994-11-09 1999-07-06 The Regents Of The University Of California Nucleic acid molecules comprising the prostate specific antigen (PSA) promoter and uses thereof
US5736524A (en) 1994-11-14 1998-04-07 Merck & Co.,. Inc. Polynucleotide tuberculosis vaccine
US5962428A (en) 1995-03-30 1999-10-05 Apollon, Inc. Compositions and methods for delivery of genetic material
NZ306767A (en) 1995-04-11 2000-03-27 Univ Johns Hopkins Method of identifying molecular interactions employing counterselection and at least two hybrid molecules or two hybrid systems
US5922687A (en) 1995-05-04 1999-07-13 Board Of Trustees Of The Leland Stanford Junior University Intracellular delivery of nucleic acids using pressure
US5585639A (en) 1995-07-27 1996-12-17 Hewlett-Packard Company Optical scanning apparatus
US5569588A (en) 1995-08-09 1996-10-29 The Regents Of The University Of California Methods for drug screening
CU22559A1 (en) 1996-01-17 1999-05-03 Ct Ingenieria Genetica Biotech EXPRESSION SYSTEM OF HETEROLOGICAL ANTIGENS IN E. COLI AS FUSION PROTEINS
WO1997033602A1 (en) 1996-03-11 1997-09-18 Cytel Corporation Peptides with increased binding affinity for hla molecules
ES2183166T3 (en) 1996-04-26 2003-03-16 Massachusetts Inst Technology TRIAL TEST FOR HYBRID TRIPLE.
US5856136A (en) 1996-07-03 1999-01-05 Incyte Pharmaceuticals, Inc. Human stem cell antigens
CA2262006A1 (en) 1996-07-26 1998-02-05 Sloan-Kettering Institute For Cancer Research Method and reagents for genetic immunization
US5925523A (en) 1996-08-23 1999-07-20 President & Fellows Of Harvard College Intraction trap assay, reagents and uses thereof
US6365797B1 (en) 1997-10-15 2002-04-02 The Regents Of The University Of California Mice models of human prostate cancer progression
ES2290964T3 (en) 1996-10-15 2008-02-16 The Regents Of The University Of California ANIMAL MODELS OF THE HUMAN PROSTATE CANCER EVOLUTION.
US5846722A (en) 1996-10-16 1998-12-08 Terrapin Technologies, Inc. System to detect small molecule/peptide interaction
US5840501A (en) 1996-10-25 1998-11-24 Bayer Corporation Determination of cPSA
AU5702298A (en) 1996-12-03 1998-06-29 Abgenix, Inc. Transgenic mammals having human Ig loci including plural VH and VK regions nd antibodies produced therefrom
US6541212B2 (en) * 1997-03-10 2003-04-01 The Regents Of The University Of California Methods for detecting prostate stem cell antigen protein
NZ337413A (en) 1997-03-10 2003-02-28 Univ California Antibodies that bind to Prostate Stem Cell Antigen (PSCA) to treat prostate cancer.
WO1998051824A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting disease of the urinary tract
US6890749B2 (en) 1997-05-15 2005-05-10 Abbott Laboratories Reagents and methods useful for detecting diseases of the prostate
US20020192659A1 (en) * 1997-09-17 2002-12-19 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
CA2303834A1 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU5922999A (en) * 1998-09-16 2000-04-03 Genentech Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6824780B1 (en) * 1999-10-29 2004-11-30 Genentech, Inc. Anti-tumor antibody compositions and methods of use
NZ518477A (en) * 1999-10-29 2004-10-29 Genentech Inc Isolated anti-prostate stem cell antigen (PSCA) antibodies that internalise upon binding to PSCA on a mammalian cell, including a PSCA-expressing tumour cell, in vivo
AU2003207708A1 (en) 2002-02-20 2003-09-09 Sirna Therapeutics, Inc. Rna interference mediated inhibition of map kinase genes
EP1499353A4 (en) * 2002-04-15 2006-04-05 Human Genome Sciences Inc Antibodies that specifically bind to tl5
US7595379B2 (en) * 2003-05-30 2009-09-29 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
US7541442B2 (en) 2003-05-30 2009-06-02 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
SI1629088T1 (en) 2003-05-30 2012-08-31 Agensys Inc Prostate stem cell antigen (psca) variants and subsequences thereof
HN2004000285A (en) * 2003-08-04 2006-04-27 Pfizer Prod Inc ANTIBODIES DIRECTED TO c-MET
EP2583981A3 (en) * 2004-05-28 2013-07-31 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
AU2006237616B9 (en) 2005-04-14 2012-12-06 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
JO3421B1 (en) 2011-06-20 2019-10-20 H Lundbeck As Method of administration of 4-((1R,3S)-6-chloro-3-phenyl-indan-1-yl)-1,2,2-trimethyl-piperazine and the salts thereof in the treatment of schizophrenia
WO2013037693A2 (en) 2011-09-15 2013-03-21 Convergent Information Technologies Gmbh System and method for the automatic generation of robot programs
US9804664B2 (en) 2014-05-27 2017-10-31 Qualcomm Incorporated Adaptive control of RF low power modes in a multi-rate wireless system using MCS value

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6258939B1 (en) * 1997-03-10 2001-07-10 The Regents Of The University Of California PSCA antibodies and hybridomas producing them
US6635468B2 (en) * 1997-09-17 2003-10-21 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same

Also Published As

Publication number Publication date
US20060029940A1 (en) 2006-02-09
KR20060021871A (en) 2006-03-08
US20130041138A1 (en) 2013-02-14
EP2319524B1 (en) 2013-08-21
US7622564B2 (en) 2009-11-24
CA2526274A1 (en) 2005-02-17
RU2005141341A (en) 2006-06-10
BRPI0410842B1 (en) 2019-09-24
CN101090729B (en) 2014-07-23
WO2005014780A3 (en) 2007-04-05
NO20055508D0 (en) 2005-11-22
SI1629088T1 (en) 2012-08-31
DK1629088T3 (en) 2012-05-07
IS8148A (en) 2005-11-24
NZ543840A (en) 2009-09-25
WO2005014780A2 (en) 2005-02-17
AU2004263823A1 (en) 2005-02-17
CA2526274C (en) 2015-12-01
EP1629088A2 (en) 2006-03-01
NO20055508L (en) 2006-02-27
IL171966A (en) 2015-09-24
CN101090729A (en) 2007-12-19
EP1629088B1 (en) 2012-01-11
CY1113044T1 (en) 2016-04-13
US8278424B2 (en) 2012-10-02
IL171966A0 (en) 2006-04-10
EP1629088A4 (en) 2008-04-02
PT1629088E (en) 2012-04-10
ZA200509606B (en) 2009-05-27
BRPI0410842B8 (en) 2021-05-25
DK2319524T3 (en) 2013-11-11
PL1629088T3 (en) 2012-06-29
ES2384622T3 (en) 2012-07-10
US20100055120A1 (en) 2010-03-04
HK1088635A1 (en) 2006-11-10
AU2008258185A1 (en) 2009-01-15
AU2008258185B2 (en) 2011-12-08
BRPI0410842A (en) 2006-07-04
NZ578591A (en) 2011-01-28
IS2900B (en) 2014-10-15
MXPA05012957A (en) 2006-06-27
JP2007525196A (en) 2007-09-06
NZ572940A (en) 2010-04-30
US20110262968A1 (en) 2011-10-27
US8853364B2 (en) 2014-10-07
EP2319524A1 (en) 2011-05-11
KR101291787B1 (en) 2013-08-07
ATE541052T1 (en) 2012-01-15

Similar Documents

Publication Publication Date Title
AU2004263823B2 (en) Prostate stem cell antigen (PSCA) variants and subsequences thereof
AU2003236553B2 (en) Nucleic acids and corresponding proteins entitled 202P5A5 useful in treatment and detection of cancer
CA2462653C (en) Nucleic acid and corresponding protein entitled 161p2f10b useful in treatment and detection of cancer
US8460881B2 (en) Nucleic acids and corresponding proteins entitled 254P1D6B useful in treatment and detection of cancer
AU2008202540A1 (en) Nucleic acid and corresponding protein entitled STEAP-1 useful in treatment and detection of cancer
AU2004235755A1 (en) Nucleic acids and corresponding proteins entitled 109P1D4 useful in treatment and detection of cancer
AU2008202217B2 (en) Nucleic acids and corresponding proteins entitled 191PAD12(b) useful in treatment and detection of cancer
CA2588564A1 (en) Nucleic acids and corresponding proteins entitled 158p3d2 useful in treatment and detection of cancer
AU2007231823B2 (en) Nucleic acids and corresponding proteins entitled 254P1D6B useful in treatment and detection of cancer
CA2503346A1 (en) Nucleic acid and corresponding protein entitled 24p4c12 useful in treatment and detection of cancer
CA2496566A1 (en) Nucleic acid and corresponding protein entitled 98p4b6 useful in treatment and detection of cancer
AU2007237184B2 (en) Nucleic acids and corresponding proteins entitled 202P5A5 useful in treatment and detection of cancer
AU2007216892A1 (en) Nucleic acids and corresponding proteins entitled 273P4B7 useful in treatment and detection of cancer

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired